study of resistance to artemisinins in plasmodium...

79
i STUDY OF RESISTANCE TO ARTEMISININS IN PLASMODIUM FALCIPARUM ISOLATES FROM KILIFI COUNTY NDUNGU DUNCAN NDEGWA A Research Thesis Submitted to the Graduate School in Partial Fulfillment for the Requirements of the Master of Science Degree in Biochemistry of Egerton University EGERTON UNIVERSITY JANUARY, 2011

Upload: buidien

Post on 11-May-2018

214 views

Category:

Documents


1 download

TRANSCRIPT

i

STUDY OF RESISTANCE TO ARTEMISININS IN PLASMODIUM FALCIPARUM

ISOLATES FROM KILIFI COUNTY

NDUNGU DUNCAN NDEGWA

A Research Thesis Submitted to the Graduate School in Partial Fulfillment for the

Requirements of the Master of Science Degree in Biochemistry of Egerton University

EGERTON UNIVERSITY

JANUARY, 2011

ii

DECLARATION AND RECOMMENDATION

DECLARATION

I declare that this thesis is my original work and has not been submitted wholly or in part

in this form or any form for a degree in this or any other university

Ndungu Duncan Ndegwa

Signature ……………….. Date ……………………

RECOMMENDATION

We wish to confirm that this research thesis has been prepared under our supervision and

have our approval to be presented for examination as per the Egerton University

Dr. A. Orina Isaac

Signature ……………………………… Date……………………………..

Egerton University, Kenya.

Dr. Steffen Borrmann

Signature ……………………………… Date……………………………..

Heidelberg University, Germany.

Dr. Maina Joseph

Signature ……………………………… Date……………………………..

Moi University, Kenya.

iii

COPYRIGHT

All rights reserved. No part of this thesis may be reproduced, stored in a retrieval system or transmitted, in any form or by any means, electronic, mechanical, photocopying or otherwise, without permission from the author or Egerton University.

© Ndungu Duncan Ndegwa 2012

iv

ACKNOWLEDGEMENT

I owe my unwavering gratitude and appreciation to the following for their great support

without which I could not have made it this far:

1. My immediate and extended family members for their financial and moral support.

2. The German Academic Exchange Services - DAAD for the in-country scholarship and

the short study fellowship to Heidelberg University, Germany.

3. Dr. Steffen Borrmann, for hosting and allowing me to work in his laboratory in

Heidelberg University, Germany.

4. My supervisors Dr. A. Orina Isaac, Dr. Steffen Borrmann and the late Dr. Maina

Joseph for their endless support.

5. The entire AG. Borrmann, for their moral and technical support: Anja Rippert for her

technical support in carrying out the project; Elise Schieck for her support in making the

statistical analysis; Not forgetting Yvonne Maier, Judith Straimer, Mamadou Tekete and

Patricia Jebet.

Above all, the almighty God and may He bless them all.

v

ABSTRACT

Malaria is a health problem and drug resistance to antimalarial poses a great challenge to

its treatment and control. Having a rapid activity against malaria parasites, artemisinins are the

front line antimalarials. However, already there are reports on reduced response to artemisinins.

Thus it’s important to monitor resistance in malaria risky areas. Merozoite surface protein (MSP)

1 and 2 genotyping has shown difference between isolates from primary and recurrent infections

but how this relates to their in vitro sensitivity is not known. The study aimed at determining

resistance of P. falciparum to artemisinins in Kilifi County. Specifically to determine: 1. in vitro

sensitivity of P. falciparum to artemisinins; 2. the difference between their in vitro activity with

that of other antimalarial drugs; 3. the existence of in vitro cross-resistance; and 4. whether there

is difference in the in vitro response to artemisinins in isolates from primary and recurrent

infection. Parasites isolates were cultured in vitro and drug response assay carried out by

subculturing them in various drug concentrations. In vitro activity was determined as the drug

concentration that inhibited 50% of parasite growth (IC50) using SYBR green I microtest.

Kruskal Wallis and Dunn’s post test were used to compare the in vitro activity of artemisinins

and that of other antimalarial drugs. Mann Whitney test was used to determine the difference in

the in vitro response of the isolates from initial and recurrent infection. Spearman correlation

analysis was used to determine existence of in vitro cross-resistance patterns between

artemisinins and other antimalarials. The mean IC50s (in nM) of Dihydroartemisinin (DHA),

Piperaquine (PQ), Lumefantrine (LMF), Chloroquine (CQ), Quinine (Q), Pyrimethamine (PYR),

Desethylamodiaquine (DE-A) and Mefloquine (MFQ) were 1.861, 30.34, 46.5, 24.29, 46.7,

18074, 37.8 and 41.7 nM respectively. There was significant difference in the in vitro activities

of the antimalarial drugs, DHA being the most active drug. There was no significant difference

in the in vitro response of the isolates from initial and recurrent malarial infections towards the

antimalarials. There was no significant correlation in the IC50s of the eight antimalarial drugs.

Therefore, artemisinins are still active in Kilifi County; more active than other standard

antimalarials and no cross-resistance was observed with other antimalarials. Consequently, as

ACTs they should be continued to be used as first line antimalarials.

vi

TABLE OF CONTENTS DECLARATION AND RECOMMENDATION .................................................................... ii

ACKNOWLEDGEMENT ...................................................................................................... iv

ABSTRACT...............................................................................................................................v

LIST OF FIGURES .............................................................................................................. viii

LIST OF TABLES .................................................................................................................. ix

LIST OF ABBREVIATIONS ...................................................................................................x

CHAPTER ONE .......................................................................................................................1

INTRODUCTION .....................................................................................................................1

1.1 Background Information ....................................................................................................1

1.1.2 Malaria Control Strategies ...........................................................................................3

1.2 Statement of the Problem ...................................................................................................5

1.3 Objectives ..........................................................................................................................6

1.3.1 General objective.........................................................................................................6

1.3.1 Specific objectives .......................................................................................................6

1.4 Hypotheses ........................................................................................................................6

1.5 Justification .......................................................................................................................7

1.6 Expected Outputs……………………………………………………………………………7

CHAPTER TWO ......................................................................................................................8

LITERATURE REVIEW .........................................................................................................8

2.1 Artemisinins ......................................................................................................................8

2.2 Mechanisms of Artemisinins’ Action .................................................................................8

2.3 Resistance to Artemisinins .................................................................................................9

2.4 In Vitro Drug Assays .........................................................................................................9

CHAPTER THREE ................................................................................................................ 12

MATERIALS AND METHODOLOGY ................................................................................ 12

3.1 Study Area. ...................................................................................................................... 12

3.2. Plasmodium falciparum Parasites Samples...................................................................... 12

3.3 Methods ........................................................................................................................... 13

3.3.1 In Vitro Culturing of P. falciparum ............................................................................ 13

3.3.1.1 Thawing of Parasites isolates..…………………………………………………...13

vii

3.3.1.2 Adaptation of Parasites into Culture.…………………………………………….14

3.3.1.3 Routine Parasite Culture.………………………………………………………...14

3.3.1.4 Splitting of Culture.……………………………………………………………...15

3.3.1.5 Giemsa Staining....……………………………………………………………….15

3.3.1.6 Parasite Freezedown.…………………………………………………………….15

3.3.1.7 Synchronization of Cultures..……………………………………………………16

3.3.2 In Vitro Drug Sensitivity Assay ................................................................................. 16

3.3.2.1 Drug Sensitivity Assay..…………………………………………………………16

3.3.2.2 Preparation of Stock Drug solution...…………………………………………….16

3.3.2.3 Preparation of the Mother Plate...………………………………………………..17

3.3.2.4 Dilution of Culture...……………………………………………………………..18

3.3.2.5 Preparation of the Working Plate...………………………………………………18

3.3.3 SYBR Green I Assay ................................................................................................. 18

3.3.3.2 Growth Inhibition Assay...……………………………………………………….19

RESULTS AND DISCUSSION .............................................................................................. 21

4.1 RESULTS ....................................................................................................................... 21

4.1.1 The In Vitro response of P. falciparum isolates to Artemisinins and other Antimalarials. .................................................................................................................... 21

4.1.2 The Difference between the In Vitro Activities of Artemisinins with that of other Antimalarial Drugs. ............................................................................................................ 26

4.1.3 The Difference in the In Vitro response to Artemisinins in P. falciparum Isolates from Primary and Recurrent Malaria infection in Kilifi County. ................................................. 29

4.1.4 The Correlation Patterns between Artemisinins and other Antimalarials .................... 34

4.2 DISCUSSION.................................................................................................................. 37

CHAPTER FIVE..................................................................................................................... 46

CONCLUSIONS ..................................................................................................................... 46

RECOMMENDATIONS ........................................................................................................ 46

REFFERENCES ..................................................................................................................... 47

viii

LIST OF FIGURES

Figure 1: Plasmodium falciparum endemicity map ......................................................................1

Figure 2: Lifecycle of P. falciparum in the host and vector ..........................................................2

Figure 3: Artemisinins .................................................................................................................8

Figure 4: The proposed mode of action of artemisinins................................................................9

Figure 5: Study site. .................................................................................................................. 12

Figure 6: Rate of in vitro resistance or susceptibility of Plasmodium falciparum to

the eight antimalarial drugs tested. .............................................................................. 25

Figure 7: Difference between the in vitro activities of artemisinins with that of other

standard antimalarial drug. .......................................................................................... 27

Figure 8: A comparison of the in vitro response of the parasite isolates from initial

infection (d0) and recurrent infection (dRC)…………………………………………..30

Figure 9: A comparison of the in vitro response of the parasite isolates from initial

infection (d0) and recurrent infection (dRC).. ............................................................. 31

Figure 10: A comparison of the in vitro response of the parasite isolates from initial

infection (d0) and recrudescent infection (dRD).. ........................................................ 32

Figure 11: A comparison of the in vitro response of the parasite isolates from initial

infection (d0) and re-infection (dRI) ........................................................................... 33

Figure 12: Correlation between the IC50s values of the eight antimalarial drugs tested.............. 35

ix

LIST OF TABLES

Table 1: Plasmodium falciparum parasites samples ................................................................... 13

Table 2: Preparation of stock drug solution ............................................................................... 17

Table 3: The In vitro response of P. falciparum isolates to antimalarials ................................... 22

Table 4: Mean IC50s of the eight antimalarial drugs tested against field isolates of

P. falciparum in Kilifi County .................................................................................... 23

Table 5: Number of resistant or susceptible parasites ................................................................. 24

Table 6: Difference between the in vitro activities of the antimalarial drugs tested .................... 28

Table 7: Correlation between the IC50s values of the eight antimalarial drugs tested ................. 36

x

LIST OF ABBREVIATIONS

ACTs: Artemisinin combination therapies

CO2 : Carbon dioxide

CQ: Chloroquine

DDT: Dichlorodiphenyltrichloroethane

DELI: Double-site enzyme-linked LDH immunodetection assay

DEA: Desethylamodiaquine

DHA: Dihydroartemisinin

DMSO: Dimethyl sulfoxide

DV: Digestive vacuole

EDTA: Ehtylenediaminetetraacetic acid

ELISA: Enzyme-linked immunosorbent assays

HCl: Hydrochloric acid

HRP- 2: Histidine-rich protein-2

KEMRI-WTRP: Kenya Medical Research institute – Wellcome Trust Research Programme

LMF: Lumefantrine

O2 : Oxygen

MHC – I: Major Histocompatibility class I

MFQ: Mefloquine

MSF: Malaria SYBR green I Fluorimetric Assay

MSP: Merozoite surface protein

NaCl: Sodium Chloride

N2 : Nitrogen

PfEMP1: P. falciparum erythrocyte membrane protein-1

pLDH: P. falciparum lactate dehydrogenase

xi

PQ: Piperaquine

PYR: Pyrimethamine

Rpm: Revolutions per minute

Q: Quinine

SP or PYR-SD: Sulphadoxine – Pyrimethamine

WHO: World health organisation

1

CHAPTER ONE

INTRODUCTION

1.1 Background Information

Malaria is a health problem in the world (Figure1) out of which 3.3 billion people are at

risk, over 300 million every year get infected and killing between 2 - 3 millions (Snow et al.,

2005). Which is a big number caused by only a single species of Plasmodium (Van Dooren et al.,

2002). Mostly it affects children less than 5 years (Breman et al., 2004; Tärning 2007) and

pregnant women. As a result of this huge number of casualty, malaria restricts development in

poor countries (Tripathi et al., 2005) due to mobilization of resources to fight it in place of other

priority needs.

In humans, malaria is caused by four species of Plasmodium; P. falciparum, P. vivax, P.

malariae and P. ovale (Day and Marsh, 1991); and P. falciparum kills more than one million

people annually (Roos et al., 2002). The tropical areas of the world provide good environment

Figure 1: Plasmodium falciparum endemicity map (Hay et al., 2009; Straimer 2009)

2

for the vectors and thus transmission of malaria, as a result the continent of Africa is the most

affected, with 60% of all malaria infections and 80% deaths (WHO, 2005). South East Asia and

South America are also highly affected (Tärning 2007).

1.1.1 Malaria Lifecycle

Figure 2: Lifecycle of P. falciparum in the host (a) and vector (b) (Menard 2005, Straimer 2009).

Malaria infection begins by an infected mosquito biting a human host. At the same time,

sporozoites from the salivary gland of the mosquito enter the bloodstream. They migrate and

within 45 (Tärning 2007) to 60 minutes (Tripathi et al., 2005) invade the liver cells, where they

multiply and develop into exo-erythrocytic schizonts (Wernsdorfer and Mc Gregor, 1988). These

schizonts burst releasing merozoites which invade erythrocytes, replicate and develop into ring

stages. Ring stages mature and develop into trophozoites. These inturn develop into erythrocytic

schizonts which bursts releasing merozoites that initiates another string of infections by invading

other erythrocytes. In the process, some merozoites develop into male or female gametocytes.

These are then taken up by a mosquito during a blood meal, and they contribute to the sexual

reproduction phase. Whereby, inside the mosquito gut the gametes fuse as a form of fertilization

resulting into a zygote which develops into oocysts. These burst and produce sporozoites that

migrate through the haemolyph to the salivary glands (Griffith et al., 2007; Tärning 2007). The

ability to cause the malaria disease is due to the high rate of multiplication which results in

3

rupturing of the erythrocytes and release of toxins. The parasite is able to modify the erythrocyte

which leads to complication of the disease whereby, by forming knobs on the erythrocytes the

parasite is able to sequester on the blood vessels avoiding clearance by the spleen (Tripathi et al.,

2005).

1.1.2 Malaria Control Strategies

The control and elimination of malaria rely on the control of the vector and management

of malaria infections. By controlling the vector it helps in reducing transmission of the disease.

The control of the vector involves the use of insecticide-treated nets, in-door and out-door

spraying. Both approaches have an impact, whereby treated nets can be maintained for long

periods and regular spraying can provide a long lasting protection (WHO, 2005). To achieve

complete vector control, is however expensive if not impractical (Miller et al., 2007; Tärning

2007).

Management of infections relies on prevention of transmission and treatment. A malaria

vaccine would prevent infection with malaria. Hence, there is need for an effective vaccine. But

this is still far from being achieved as there is currently no any functional and acceptable

vaccine. Of the many vaccines that have been presented only a few have passed the early clinical

trials (Aide et al., 2007; Snounou and Renia, 2007) and many are yet to be presented due to the

availability of P. falciparum genome sequence (Gardner et al., 2002).

The prevention of infection with malaria is further weakened by the immune evasion

challenges the parasite poses to the immune system preventing parasite clearance consequently

resulting in continual malarial infections:

The sporozites avoid immune detection and destruction as once they are in the

bloodstream they move quickly to the liver where they change into merozoites, thus they don’t

become exposed to antibodies and T cells respectively (Taubes, 2000).

The erythrocytes lack antigen presenting receptors i.e. MHC – I receptors as such, the

parasites become hidden from T cells (Janeway et al., 2005).

The parasite modifies erythrocytes by making them rigid, interfering with transport

mechanisms and PfEMP1 encoded by var genes is deposited into the membrane forming Knobs.

4

Through the knobs infected erythrocytes attach to walls of the blood vessels preventing their

destruction by the spleen. These knobs also result in infected erythrocytes binding to uninfected

ones, forming a complex that leads to blockage of blood vessels. These modifications result in

complication of malaria. PfEMP1 is involved in antigenic variation whereby as a result of the

immune pressure the parasite changes the sequence of var genes expressed resulting in immune

evasion (Hastings et al., 2004). Additionally, PfEMP1 can attach to dendritic cells, interfering

with the immune response process (Craig and Scherf, 2001). To avoid destruction gametocytes

remain inactive while in the human host (Taubes, 2000).

With the above complications in the control and management of malarial, an alternative

to avoiding the malaria burden is the use of effective, safe and cheap antimalarials (Tärning

2007). Treatment of malaria has always relied on antimalarials such as chloroquine,

sulphadoxine-pyrimethamine etc.

After chloroquine was discovered, its use together with the insecticide DDT boosted the

fight and elimination of malaria in the western world. However, in developing countries political

instabilities, shortage of funds among other factors hindered this fight, resulting in resistance to

CQ and SP (Wellems and Plowe, 2001; Gregson and Plowe, 2005). Antimalarial drug resistance

later spread to MFQ or Q (Wongsrichanalai et. al., 2002) worsening the malaria burden.

The wide geographic distribution of P. falciparum (Tärning 2007) influences the

dynamics of antimalarial drug resistance (Hastings and Watkins, 2005) greatly inhibiting the

fight against malaria. There are low and high transmission areas e.g. South east of Asia and

Africa respectively. People in these areas thus experience different infectious bites per person per

year e.g. 2 and up to 1500 respectively (Hay et al., 2000). Consequently, people in low

transmission areas develop malaria symptoms as opposed to those in the high. This is a result of

the latter developing partial immunity to malaria but which wanes out when they migrate to low

transmission areas. Thus, in low transmission areas there is frequent treatment with antimalarials

than from the high (Breman et al., 2004). As a result there is a lot of drug pressure in the low

transmission areas which selects resistant parasites that inturn spread to the high transmission

areas e.g. CQ and SP first emerged in south east asia and spread to africa. These drugs were

affordable and easily available (Touré et al., 2008; Lim and McFadden, 2010), worsening the

5

malaria fight (Greenwood et al., 2005) thus necessitating new and different antimalarials (White,

2004).

ACTs are effective against parasites resistant to other common antimalarials and they are

recommended by the WHO as the front-line drugs. Indeed in combination with other control

strategies they are proving useful (Feachem and Sabot, 2008; Eastman and Fidock, 2009).

Hence, maintaining the optimism in malaria elimination (Kaddouri et al., 2006).

The basis of ACTs is, since resistance results from mutations in the target or transport

proteins, it would be hard for a parasite to become resistance at the same time to two drugs that

act differently in comparison to a single drug usage (White, 1999; Nosten and White, 2007; Cui

and Su, 2009). Examples of ACTs include; Artemether–lumefantrine and dihydroartemisinin–

Piperaquine.

However, there are accounts of reduced response to these antimalarials in the South East

of Asia (Cui and Su, 2009; Dondorp et al., 2009; Leah et al., 2009). Hence generating fears

towards spread of this resistance and erosion of their utility (Wootton et al., 2002; Roper et al.,

2004). Assessment of P. falciparum drug susceptibility in Kilifi is thus necessary to sustain

health recommendations for malaria treatment and prophylaxis in Kenya and other countries

where malaria is endemic (Kaddouri et al., 2006).

During the time course of malaria tropica infection different clones of P. falciparum

parasites have been isolated and differentiated by genotyping of the MSP 1 and 2. These result in

primary and recurrent infections. Primary referring to the initial infection and recurrent to

subsequent infections, which is associated with two different parasite clones; recrudescent,

which are similar genotypically to those of the primary infection and new different parasites.

Thus, does this difference translate into differences in drug response or polymorphisms in

Pfatpase6 and Pfmdr1 genes associated in artemisinin resistance?

1.2 Statement of the Problem

Antimalarial drug resistance is a great limitation in the fight against malaria, since there

is a high rate of resistance to antimalarials developing and spreading. The lack of a new and

different antimalarial worsens the situation. As a result the possibility of parasites developing

resistance to artemisinins threatens the usefulness of ACTs. Making it mandatory to monitor

6

artemisinins susceptibility and hence researching more on this, hence, the need to determine

artemisinins resistance in endemic areas such as Kilifi.

Additionally, through MSP 1 and 2 genotyping there is difference between parasite

clones from initial and recurrent malaria infection but how this relates to their in vitro sensitivity

toward artemisinins needs to be determined.

1.3 Objectives

1.3.1 General objective

To determine resistance of P. falciparum to artemisinins in Kilifi County

1.3.1 Specific objectives

1. To determine in vitro response of P. falciparum isolates to artemisinins and to establish

the sensitivity level of artemisinins in Kilifi County.

2. To determine difference between the in vitro activity of artemisinins with that of other

antimalarial drugs.

3. To determine whether there is a difference in the in vitro response to artemisinins in P.

falciparum isolates from primary and recurrent malaria infection in Kilifi County.

4. To determine existence of in vitro cross-resistance between artemisinins and other

antimalarials.

1.4 Hypotheses

1. P. falciparum isolates from Kilifi County have reduced in vitro response to

artemisinins?

2. There is difference between the in vitro activity of artemisinins and other antimalarials

towards P. falciparum isolates from Kilifi County?

3. There is difference in the in vitro sensitivity towards artemisinins in P. falciparum

isolates from primary and recurrent malaria infection in Kilifi County?

4. There is in vitro cross resistance between artemisinins and other antimalarials towards

P. falciparum isolates from Kilifi County?

7

1.5 Justification

Guiding principles for malaria treatment rely on in vitro drug tests to provide information

on the susceptibility profiles of parasites. In this way the study will demonstrate the status on P.

falciparum response to artemisinins in Kilifi County, generating background information which

can be used to asses and improve treatment guidelines.

1.6 Expected Outputs

1. The sensitivity level of artemisinins in Kilifi County will be established to inform

policy on treatment programs.

2. Master of Science Degree in Biochemistry.

3. Publication in peer reviewed journals.

8

CHAPTER TWO

LITERATURE REVIEW

2.1 Artemisinins

The artemisinins is a product of research by Chinese scientists (Klayman, 1985; Cui and

Su, 2009). Besides malaria, they are able to treat schistosomiasis and cancer (Krishna et al.

2008). Their activity against malaria is due to an endoperoxide bridge they posses, and can kill

parasites in several minutes (Woodrow et al., 2005; White, 2008; Cui and Su, 2009). They are

extracted from Artemisia annua and then modified to other derivatives due to their instability and

insolubility (Touré et al., 2008). Alternatively, they can also be obtained through genetic

engineering using the yeast cell (Ro et al., 2006; Eastman and Fidock 2009).

Figure 3: Artemisinins (Cui and Su, 2009). Artemisinins are broken down to DHA, their active component (Wongsrichanalai et al., 1999; Schlitzer 2008; Mwai et al. ., 2009).

2.2 Mechanisms of Artemisinins’ Action

The mode of action of artemisinins is not yet well understood and there are two possible

ways how this happens (Figure 4): either, the endoperoxide bridge is reduced by haem iron in the

digestive vacuole (Hartwig et al., 2009) generating toxic radicals (Krungkrai and Yuthavong,

1987; Asawamahasakda et al., 1994; Kannan et al., 2005) or the bridge is reduced by iron-

sulphur redox centres (Wu, 2002).

9

Figure 4: The proposed mode of action of artemisinins: The Endoperoxide Bridge is the active

component of artemisinins which undergoes reduction to produce radicals whose target is not yet

clear (Schlitzer 2008).

2.3 Resistance to Artemisinins

Artemisinin resistance has been demonstrated in P. falciparum lab strain, F32-ART

(Witkowski et al., 2009). And despite the lack of demonstrable resistance in isolates from

patients, there are reports of reduced response to these antimalarials in different parts of the

world, which include: western Thailand (Luxemburger et al., 1998), Cambodia (Denis et al.,

2006; Vijaykadga et al., 2006; Cui and Su, 2009), India (Gogtay et al., 2000), Sierra Leone

(Sahr et al., 2001), Nigeria and Madagascar (Oduola et al., 1992; Randrianarivelojosia et al.,

2001), and Yunnan province of western China (Yang et al., 2003), French Guiana and Senegal

(Jambou et al., 2005).

As we await more confirmation on the same issue, these are proof of resistance to

artemisinins in P. falciparum. This is a threat to the fight against malaria (O'Neill et al.. 2010),

consequently, the need to determine artemisinins resistance in endemic areas such as Kilifi.

2.4 In Vitro Drug Assays

Techniques for assessing artemisinins resistance include in vivo and in vitro assays. In

vitro enable determination of inherent antimalarial drug resistance, they are advantageous in

being cheap and from free from host factors (Bacon et al., 2007), While the in vivo, are

applicable in determining clinical resistance (Noedl et al., 2004). In vitro assays include:

10

1. The WHO microtest, in which results are, assessed microscopically, a time-consuming

process.

2. Isotopic microtest; which requires high parasitemia (Druile et al., 2001), its expensive

and dangerous involving radioactive materials.

3. Molecular marker determination which sometimes doesn’t connect to in vitro drug

resistance (Bacon et al., 2007).

4. Colorimetric assays based on biochemical reactions resulting in colour development.

This is available in various types:

a) Fluorescent-based techniques that use fluorescent dyes e.g. SYBR green I that

intercalate with DNA (Johnson et al., 2007). It is specific to the erythrocytic stage since

erythrocytes lack Nucleic acids (Bennett et al., 2004, Johnson et al., 2007).

b) Sensitive quantification of major parasite proteins by sandwich ELISA based on

labeled primary and secondary antibodies directed to distinct epitopes of the same antigen

giving greater sensitivity (Druilhe et al., 2001). The antibodies are for antigen capture

and identification respectively.

Antigens targeted are either only found in a specific Plasmodium species or are found in

all of them. Those specific to P. falciparum are HRP-2 and pLDH used in HRP2 ELISA (Noedl

et al., 2004, 2005) and DELI (Druilhe et al., 2001) respectively. On the contrary pLDH and

aldolase are found in all species. Aldolase is used in sandwich aldolase ELISA (Murray and

Bennett 2009).

The discrimination on the type of assay to use involves the time taken to culture parasite.

Hypoxanthine, pLDH and SYBR green I assay needs 48 or 72 hour. HRPII assay needs 72-hour

inhibiting its application in assays requiring shorter times (Bacon et al., 2007). Additionally,

mutations in the targeted antigens can lead to wrong results (Murray and Bennett, 2009).

Aldolase based tests are applicable in a parasitemia of atleast 0.03% (Tritten et al., 2009).

While in the DELI, a parsitemia of atleast 0.005% (Druilhe et al., 2001).

11

Even though ELISA-based techniques are precise and consistent, they are however,

costly and tiresome. On the other hand, fluorescence-based techniques provide a less costly and

fast approach.

The SYRB green I assay for example: Is performed on one plate consuming fewer

reagents in comparison to the standard isotopic microtest. It’s carried out in a single process. The

SYBR green dye itself is stable. It doesn’t use any antibodies. It is less costly (Smilkstein et al.,

2004; Bacon et al., 2007; Johnson et al., 2007). Thus MSF assay will be used.

12

CHAPTER THREE

MATERIALS AND METHODOLOGY

3.1 Study Area.

The study site is Pingilikani (Figure 5) which is under a partnership involving Heidelberg

University, Germany and the KEMRI-WTRP Kilifi. It is about 20 km away southwards of

Kilifi. It experiences two rainy seasons, April - June and November – December, during which

malaria transmission maximizes (Straimer 2009). The inoculation rate is about 22 to 53 infective

bites per person per year (Mbogo et al., 2003). Artemether-lumefantrine is the current front-line

drug replacing amodiaquine which has been in use since 2003 (Sasi et al., 2009).

Figure 5: Study site: The figure on the left is a map of Kilifi County, Kenya. And the right one is

Pingilikani dispensary (Straimer 2009).

3.2. Plasmodium falciparum Parasites Samples

The samples for the study were randomly selected from a huge collection of samples

collected from Pingilikani between 2006 – 2008 in a study that was determining safety and

efficacy of Artekin in African children, among which Kilifi was one of the study site.

During the study, the parasites were isolated from the patients during the initial day of the

study (This was the day zero- d0) and subsequently during the follow up days of the study in

13

patients presenting with recurrent parasitemia (This were named according to the day of parasite

isolation).

To differentiate the P. falciparum isolates whether they are the same or different ones,

isolates from patients with recurrent parasitemia were MSP 1 / 2 genotyped; those with similar

sequences as isolates from day zero were referred to as recrudescent and those that were

different; re-infection. From this pool of isolates from day zero and day of recurrence the

following 8 isolates for this study were randomly selected.

Table 1: Plasmodium falciparum parasites samples

Day Zero Recurrence Remarks

27d0 27d21 Recrudescent

171d0 171d21 Recrudescent

115d0 115d49 Re-infection

191d0 191d54 Re-infection

The laboratory strain 3D7 was used as the control.

3.3 Methods

3.3.1 In Vitro Culturing of P. falciparum

In vitro culturing of P. falciparum was done according to Trager and Jensen (Jensen

2002) with few modifications.

3.3.1.1 Thawing of Parasites Isolates

To prepare frozen parasites for culture, cryovials stored in liquid nitrogen were

transferred into - 800C and left to thaw overnight. Then after 12-24 hours they were removed

and thawing was done by rapidly rolling the cryovials between two gloved hands. Then 200µl of

12% NaCl was added slowly to the vial and mixed by shaking gently. The mixture was

transferred to a 50 ml falcon tube. Nine millimeters of 1.6% NaCl was added drop by drop for

the first 3 ml and mixed. This was then centrifuged at 1800 rpm for 3 minutes. The supernatant

was then carefully removed by drawing it out with Pasteur pipettes connected to the vacuum

pump and this was replaced by dropwise addition of 7 ml of 0.9% NaCl and 0.2% glucose with

14

gentle agitation. This was then centrifuged at 1800 rpm for 3 minutes and the supernatant

removed as above. Then the parasite pellet was introduced directly into culture as described

below.

3.3.1.2 Adaptation of Parasites into Culture

P. falciparum culture medium was prepared by mixing 500 ml of RPMI 1640

(supplemented with L glutamine and HEPES), 250µl gentamicin, 10ml human AB serum, 5ml

hypoxanthine and 50ml Albumax II. The solution was then filtered through 0.22µ Millipore filter

connected to a vacuum pump. This was then stored at 40C.

Under sterile conditions and working under a culture hood, 10 ml of culture medium was

pipetted into the above parasite isolate then mixed by gently pipetting the mixture up and down.

This was then transferred into a T25 culture flask and 1 µl of fresh uninfected erythrocytes

pipetted into the solution and mixed again. The flask was placed in an incubator and the parasites

were incubated at 370C, in constant flowing gas mixture of 90% N2, 5% CO2 and 5% O2. The

culture media was changed after every two days and the parasitemia was checked daily by

giemsa staining as described below. When at least a single parasite was spotted the culture was

split into two portions and one portion maintained in culture and the other frozen down as

described below (Section 3.3.1.4 and 3.3.1.6 respectively);

Also after splitting of the culture to encourage faster growth the remaining portion was

transferred to T75 culture flask and 1.5 ml of fresh erythrocytes and 30 ml of fresh culture

medium was added; in all maintaining a hematocrit of 5% and a parasitemia of less than 1%. The

parasites were then maintained in culture with regular media change after two days and daily

monitoring of the parasitemia.

3.3.1.3 Routine Parasite Culture

Routine parasite culture refers to the continued culture of parasites after they had been

adapted to culture. It was done by maintaining the parasites in culture for a longer period 1 to 3

weeks through regular media change after two days and daily monitoring of the parasitemia. This

was continued until the parasites attained a two, three or four folds multiplication of the

parasitemia; which is regarded as the normal parasite cycle. During this whole period (2-3

weeks), the parasitemia was not allowed to exceed 5%, otherwise it was splitted as described

15

below (some portion being used for parasite freeze down, DNA extraction or stored as a parasite

pellet for other studies). In cases where there was a mixture of trophozoites and ring stages the

culture was made synchronous by sorbitol synchronization as described below (Section 3.3.1.7).

3.3.1.4 Splitting of Culture

Splitting of the parasites was done by mixing the culture by gentle swirling and then

using a pipette and accu-jet: In T25 culture flasks 1, 2 and utmost 3 ml of the solution was

removed and the rest transferred to 15 ml falcon tube (for freeze down). The removed solution

was returned to the same culture flask or a new one, and then 500µl of fresh erythrocytes and

10ml of fresh culture medium was added.

In T75 culture flasks 5 or 10 ml of the solution was removed and the rest transferred to

50ml falcon tube (for freeze down). The removed solution was returned to the same culture flask

or a new one, and then 1.5ml of fresh erythrocytes and 30 ml of fresh culture medium was added.

The solution was mixed gently then returned to culture.

3.3.1.5 Giemsa Staining

From the culture solution and using a 1ul pipette a drop of blood was taken and smeared

on a clean glass slide and air dried. This was then fixed by immersion in methanol for a minute

and then air-dried; then immersed for 10–30 minutes in Giemsa solution, washed under running

water and then dried. This was then examined on a microscope at 100 objectives with immersion

oil by quickly scanning through the slide and choosing an area where the red blood cells were

uniformly distributed (with no overlapping cells). One thousand erythrocytes were counted

randomly in different fields while partially filled fields were avoided. Also infected erythrocytes

were counted and they were expressed per a thousand of the uninfected cells to give the

parasitemia.

3.3.1.6 Parasite Freezedown

During parasite adaptation when at least a single parasite was spotted and the culture was

split into two portions, one portion was maintained in culture and the other frozen down by

centrifuging the culture solution at 2100 rpm for 3 minutes at 200C to obtain an erythrocyte

pellet. Then the supernatant was carefully sucked out using the vacuum pump. The volume of the

packed cells was estimated and depending on the volume, 500µl or less was transferred to a

16

cryovial, labeled with the parasite’s code and date. Then equal volume of the freezing solution (3

% sorbitol; 0.65 % NaCl and 28 % glycerol) was added followed with a gentle mixing and then

the cryovial was frozen down immediately at – 800C at least for one day and then transferred to

liquid nitrogen for long-term storage.

3.3.1.7 Synchronization of Cultures

Asynchronous culture was split as described above (Section 3.3.1.4), one portion returned

to culture and the larger portion was centrifuged at 2100 rpm for 3 minutes at 200C to precipitate

the erythrocytes. The supernatant was removed and then 10ml of sterilized 5% D-sorbitol added

to the precipitate. This was incubated in a waterbath for 10 minutes, and then centrifuged again

as later. Parasitemia was determined and the pellets returned to culture. The procedure was

repeated after one cycle (48 hours) and once a week for 2 weeks.

3.3.2 In Vitro Drug Sensitivity Assay

In vitro drug sensitivity assay was done as described in Druilhe et al. (2001)

3.3.2.1 Drug sensitivity Assay

The sensitivity of the isolates was determined for Artemisinins as DHA (this was the test

compound and it is the active compound in Artemisinins), LMF, MFQ, PQ (Because these are

used with artemisinins as ACTs), CQ, PYR, Q and DEA. The non artemisinin drugs were for

comparison and cross resistance determination.

3.3.2.2 Preparation of Stock Drug Solution

PQ, LMF, Q and DEA were dissolved in 90% methanol plus 10% HCl; DHA in 70%

ethanol; PYR in DMSO; MFQ in 99% methanol and 1% acetic acid and then CQ in distilled

water, as shown in the following table. These solutions were then diluted by culture media to

obtain a working solution. The solutions were covered with aluminum paper foil since the drugs

are light sensitive. PYR and DHA were prepared immediately when they were being used due to

their high light sensitivity and unstable nature.

17

Table 2: Preparation of stock drug solution

Compound Solvent Stock

Solution

(mg/ml)

Dilution

Factor

Working

Solution

(mg/ml)

1st

concentratio

n (ng/ml)

PQ 90% Meth

10% HCL

5 x 100 0.05 1851.85

LMF 90% Meth

10% HCL

1 x 10 0.10 3703.70

DHA 70% ethanol 1 x 200 0.005 185.18

CQ Distilled

water

5 x 100 0.05

1851.85

Q 90% Meth

10% HCL

2 x 10 0.20 7407.41

PYR DMSO 5 x 10 0.50

18518.52

DEA 90% Meth

10% HCL

5 x 100 0.05 925.925

MFQ 99% Meth

1% HCL

Acetic acid

5 x 100 0.05 1851.85

NB: Drugs were dissolved into respective solvents to obtain stock solutions. These were then

diluted with culture medium to obtain working solution. This working solution when added in a

threefold dilution to the mother plate (Section 3.3.2.3) resulted in X27 dilution of the drug from

the second well to the last well, giving the indicated first drug concentration on the second well.

3.3.2.3 Preparation of the Mother Plate

The microtiter plate that was used consisted of 96 flat-bottom wells, arranged in a matrix

of eight rows (A through H) and 12 columns (1 through 12). Two mother plates were prepared

each containing four drugs in duplicate rows, successively following the order shown in the

above table. Two hundred microliters of culture medium was then added to each well. From the

18

working drug solutions 100µl of last stock drug solution was added to each well in the second

column. These were then mixed by gentle up and down pipetting. Then 100µl of the mixture was

transferred to each adjacent well in the third column. This was also transferred to the wells in the

second column and so on until the last column, from which 100 µl of the mixture was removed.

3.3.2.4 Dilution of Culture

Dilution of culture was performed by first determining the parasitemia as described in

Section 3.3.1.5 then diluting it to 1% by first spinning down the culture solution to obtain an

erythrocyte pellet as described below. The supernatant was removed and the infected

erythrocytes mixed with uninfected ones in a ratio according to the determined parasitemia to

obtain a total count of 600µl erythrocytes. Then 30ml of media was added to the cells, in total

getting a parasitemia of 1% and a hematocrit of 2%.

Note: For IC50, lab strain parasitemia was maintained at 0.5 to 1%, mostly less than 1% this is

because they grew very fast i.e. they are culture adapted. Thus the dilution was made in 1:4 (1+3

parts of infected and uninfected erythrocytes, respectively). For field isolates, the parasitemia

was maintained at 1%, because they grow very slowly.

3.3.2.5 Preparation of the Working Plate

One hundred microliters of the culture solution (diluted to 1% parasitemia and 2%

hematocrit as described in section 3.3.2.4) was added to each well except the last three wells in

the first column. These three wells were loaded with 100µl of uninfected erythrocytes acting as

negative controls. Then 12.5 µl of the drug solution from the mother plate was added to the

working plate, transferring the contents from wells in one column to the similar wells in the other

column, respectively taking care that each well was correctly matched. The first five wells in the

first column contained no drug solution and these acted as the positive controls. The plates were

then incubated for 72 hours.

3.3.3 SYBR Green I Assay

SYBR Green I Assay was done according to Smilkstein et al. (2004). This was done to

determine growth inhibition, whereby SYBR green dye intercalates with double stranded DNA

and fluoresces (Johnson et al., 2007). As such, positive control wells will give 100%

19

fluorescence due to complete growth, while test wells will yield reduced fluorescence due to

reduced growth of the parasites.

3.3.3.1 Growth Inhibition Assay

MSF lysis buffer was prepared by mixing 1 L cell culture water with 2.423 g of Tris base.

This was then dissolved completely using a magnetic stirrer. Then the pH was adjusted to 7.5

using concentrated HCl. To this solution 10 ml 0.5 M EDTA was then added. Then 80 mg of

saponin was added, followed with 0.8 ml Triton X-100. Then the solution was mixed thoroughly,

avoiding the production of bubbles. This was then Vacuum filtered to remove particulate matter

and stored at Room Temperature.

After 72 hours of growth, 100µl of SYBR Green I in lysis buffer (2.2 µl of SYBR Green

I/11ml of lysis buffer (Tris HCl, EDTA, Triton X-100 and Saponin)) was added to each well and

the contents were mixed until no visible erythrocyte sediment remained. After 1 h of incubation

in the dark at room temperature, fluorescence was measured with Fluostar optima fluorescence

multiwell plate reader with excitation and emission wavelength bands centered at 485 and 530

nm, respectively, and a gain setting equal to 46.

3.3.4 Data Analysis

In vitro activity was assessed as the drug concentration that inhibited 50% of parasite

growth (IC50). Data was collected as fluorescence units and matching drug concentrations in

nano Moles (nM).

a) To investigate in vitro response of P. falciparum isolates from Kilifi County to

artemisinins: IC50 (ng/ml) obtained after incubation times of 48 hours was determined

using excel sheets (Microsoft corporation, USA) based on non linear regression model.

These were then converted to nM. Parasite isolates were identified as resistant or

susceptible based on IC50 cutoff of reference IC50s: DHA >10.5 nM (Bruno et al.,

2010), Q > 800 nM (Ndong et al., 2003) (Bruno P. et al., 2010), MFQ >30 nM (Ndong et

al., 2003)(Bruno P. et al., 2010), LMF >150 nM, DEA >80 nM, 100 nM (Kaddouri et al.,

2006), PYR >2,000 nM (Touré et al., 2008; Cui and Su, 2009) and PQ > 100nM

(Leonardo and Pascal, 2003; Mwai et al., 2009).

20

b) To determine difference between the in vitro activity of artemisinins with that of

other standard antimalarial drugs. The above calculated IC50s for each drug for the

samples were compared by Kruskal Wallis test and Dunn’s post test. The level of

significance was set at a P value of <0.05.

c) To determine whether there is a difference in the in vitro response to artemisinins

in P. falciparum isolates from primary and recurrent malaria infection in Kilifi

County. Mean IC50s for each drug for each group of the samples were compared by

Mann Whitney test. The level of significance was set at a P value of <0.05.

d) To determine existence of in vitro cross-resistance patterns between artemisinins

and other standard antimalarials: Correlation analysis of the above calculated IC50s

for all drugs for the samples was performed using Spearman corelation analysis. The

statistical analysis was carried out using Prism 4.0 for windows (Graphpad software Inc.

CA).

21

CHAPTER FOUR

RESULTS AND DISCUSSION

4.1 RESULTS

4.1.1 The In Vitro response of P. falciparum isolates to Artemisinins and other

Antimalarials.

Eight field isolates of P. falciparum were adapted for long-term culture and their in

vitro drug chemosensitivity profiles analyzed (Table 3). Laboratory strain 3D7 was used as the

control. The mean IC50s of PQ, LMF, DHA, CQ, Q, PYR, DE-A and MFQ were 30.34 nM

(95% CI 26.96-33.72nM), 46.5nM (95% CI 31.40-61.60nM), 1.861 nM (95% CI 1.489-2.233

nM), 24.29 nM (95% CI 13.2-35.46 nM), 46.7 nM (95% CI 36.78-56.62 nM), 18074 nM (95%

CI 9483-26665), 37.8 nM (95% CI 19.46-56.14nM) and 41.7 nM (95% CI 23.64-59.75 nM)

respectively (Table 4).

No isolate was resistant to PQ, LMF and DHA. Also considering the internationally used

IC50 cut off of > 100 nM all (8) isolates were sensitive to CQ, but considering the IC50 cutoff of

> 25 nM recommended for the study site (Mwai et al., 2009) one (1/8) isolate was resistant to

CQ. No isolate was resistant to Q considering both the high IC50 cutoff value of > 800 nM and

lower > 500 nM (Okombo et al., 2010). All (8) isolates were resistant to PYR. One (1/8) isolate

was resistant to DE-A. Five (5/8) isolates were resistant to MFQ (Table 5 and Figure 6).

22

Table 3: The In vitro response of P. falciparum isolates to antimalarials

Drugs tested

PQ LMF DHA CQ Q PYR DE-A MFQ

Control

3D7 33.03 70.31 2.04 9.31 49.43 20.58 83.07 70.95

Isolates

27d0 24.05 36.01 1.58 57.09 68.70 5347.00 38.06 53.06

27d21 34.04 53.02 2.62 22.39 51.28 29295.00 50.61 66.30

171d0 30.81 68.35 2.06 18.54 38.05 23301.00 85.83 54.41

171d21 33.58 58.58 2.38 19.15 31.85 33452.00 27.44 65.28

115d0 32.25 68.85 1.54 18.17 47.43 18168.00 19.30 41.70

115d49 24.44 35.48 1.76 21.56 56.03 17468.00 30.46 24.44

191d0 29.81 21.73 1.38 20.10 37.70 11463.00 18.81 15.33

191d54 33.77 29.99 1.57 17.35 42.58 6097.00 31.90 13.04

The in vitro response of field isolates of P. falciparum from Kilifi County to eight antimalarials expressed as IC50 in nM. The isolates are named using the number of patient it was isolated from and the day of isolation.

23

Table 4: Mean IC50s of the eight antimalarial drugs tested against field isolates of P. falciparum in Kilifi County

Drugs No. of isolates Mean IC50 95% Confidence Range (nM)

tested nM interval (nM) Minimum Maximum

PQ 8 30.34 26.96-33.72 24.05 34.04

LMF 8 46.50 31.40-61.60 21.73 68.85

DHA 8 1.86 1.49-2.23 1.38 2.62

CQ 8 24.29 13.20-35.46 17.35 57.09

Q 8 46.70 36.78-56.62 31.85 68.70

PYR 8 18074.00 9483.00-26665.0 5347.00 33452.00

DE-A 8 37.80 19.46-56.14 18.81 85.83

MFQ 8 41.7 0 23.64-59.75 13.04 66.30

24

Table 5: Number of resistant or susceptible parasites

PQ LMF DHA CQ CQ Q Q PYR DE-A MFQ

CUTOFF >100 >150 >10.5 >100 >25 >800 >500 >2,000 >80 >30

Total 8 8 8 8 8 8 8 8 8 8

Susceptible 8 8 8 8 7 8 8 0 7 3

Resistant 0 0 0 0 1 0 0 8 1 5

Parasites are determined to be sensitive or resistant based on their IC50 value to a particular drug being lower or upper than the cutoff

value respectively. The IC50 cutoff values are in nM. No isolate was resistant to PQ, LMF, DHA, CQ (>100 nM) and Q. One isolates

was resistant to CQ considering the >25nM recommended for the study site, five to MFQ and all were resistant to PYR.

25

Figure 6: Rate of in vitro resistance or susceptibility of Plasmodium falciparum to the eight antimalarial drugs tested.

26

4.1.2 The Difference between the In Vitro Activities of Artemisinins with that of other

Antimalarial Drugs.

There was significant difference (P< 0.0001) in the in vitro activities of the eight

antimalarial drugs towards the isolates (Kruskal-Wallis analysis, P<0.05) (Figure 7). As

expected, DHA was the most active drug, followed by CQ, PQ, DE-A, MFQ, LMF, Q and lastly

PYR (Table 5). Specifically there was significant difference between the in vitro activity of DHA

compared to LMF, CQ, Q, PYR, and MFQ. But there was no significant difference when

compared to PQ and DEA (Table 6).

27

Figure 7: Difference between the in vitro activities of artemisinins with that of other standard antimalarial drugs: Kruskal-Wallis

analysis of the difference between the IC50s of the eight antimalarial drugs tested. There was significant difference; P< 0.0001 with

DHA being the most active.

28

Table 6: Difference between the in vitro activities of the antimalarial drugs tested; Dunn’s post test, P<0.05

Drug pair Summary Drug pair Summary

PQ-LMF ns DHA-Q **

PQ-DHA ns DHA-PYR ***

PQ-CQ ns DHA-DEA ns

PQ-Q ns DHA-MFQ *

PQ-PYR * CQ-Q ns

PQ-DEA ns CQ-PYR ***

PQ-MFQ ns CQ-DEA ns

LMF-DHA ** CQ-MFQ ns

LMF-CQ ns Q-PYR ns

LMF-Q ns Q-DEA ns

LMF-PYR ns Q-MFQ ns

LMF-DEA ns PYR-DEA *

LMF-MFQ ns PYR-MFQ ns

DHA-CQ ns DEA-MFQ ns

Dunn's Multiple Comparison Test; post test of the Kruskal-Wallis analysis of the difference between the IC50s of the eight antimalarial drugs tested. ns; non-significant = P > 0.05.*; significant = P < 0.05. **; very significant = P < 0.01, ***; highly significant = P < 0.001.

29

4.1.3 The Difference in the In Vitro response to Artemisinins in P. falciparum Isolates from

Primary and Recurrent Malaria infection in Kilifi County.

There was no significant difference (P = 0.9591) in the in vitro response of the isolates

from initial and recurrent malarial (both recrudescent and re-infections) infections towards

dihydroartemisinin and the other antimalarial drugs tested (Mann Whitney test, P <0.05) (Figure

8, 9, 10 and 11).

30

Figure 8: A comparison of the in vitro response of the parasite isolates from initial infection (d0) and recurrent infection (dRC).

31

Figure 9: A comparison of the in vitro response of the parasite isolates from initial infection (d0) and recurrent infection (dRC). There

was non-significance difference (P = 0.9591). Mann Whitney test P <0.05.

32

Figure 10: A comparison of the in vitro response of the parasite isolates from initial infection (d0) and recrudescent infection (dRD).

There was non-significance difference (P = 0.9591). Mann Whitney test P <0.05.

33

Figure 11: A comparison of the in vitro response of the parasite isolates from initial infection (d0) and re-infection (dRI). There was

non-significance difference (P = 0.9591). Mann Whitney test P <0.05.

34

4.1.4 The Correlation Patterns between Artemisinins and other Antimalarials

There was no significant correlation in the IC50s of the eight antimalarial drugs towards

the isolates (Spearman correlation analysis, P<0.05) (Figure 12 and Table 7).

35

Figure 12: Correlation between the IC50s values of the eight antimalarial drugs tested.

36

Table 7: Correlation between the IC50s values of the eight antimalarial drugs tested; Spearman correlation, P<0.05

Except between DHA vs MFQ and PYR vs MFQ there was no significant correlation in the IC50s of the eight antimalarial drugs towards the isolates. But the DHA vs MFQ and PYR vs MFQ correlation is highly unlikely (Spearman correlation analysis, P<0.05). ns; non-significant = P > 0.05.*; significant = P < 0.05.

Drug pair r2 P Drug pair r2 P

DHA-MFQ 0.83 * PQ-MFQ 0.29 ns

PYR-MFQ 0.74 * LMF-DEA 0.24 ns

DHA-PYR 0.69 ns PQ-LMF 0.21 ns

DHA-DEA 0.64 ns CQ-DEA 0.21 ns

LMF-MFQ 0.64 ns PQ-DEA 0.14 ns

LMF-PYR 0.62 ns PYR-DEA 0.10 ns

CQ-Q 0.55 ns DHA-Q 0.05 ns

PQ-PYR 0.55 ns Q-MFQ 0.00

DEA-MFQ 0.48 ns LMF-Q -0.05 ns

PQ-DHA 0.38 ns CQ-PYR -0.10 ns

LMF-DHA 0.38 ns LMF-CQ -0.21 ns

CQ-MFQ 0.40 ns PQ-Q -0.36 ns

Q-DEA 0.36 ns PQ-CQ -0.43 ns

DHA-CQ 0.33 ns Q-PYR -0.45 ns

37

4.2 DISCUSSION

As a result of P. falciparum developing resistance to common antimalarials such as CQ

(Le Bras et al., 2006) there is need of a novel antimalarial to boost the fight against malaria.

ACTs are currently recommended as the front line dugs (Nosten and White 2007; Eastman and

Fidock 2009). The partner drugs used in the combination protect each other in terms of resistance

(White 2001). However, the parasite as stubborn as it is has started developing resistance (Noedl

et al., 2008; Dondorp et al., 2009; Rogers et al., 2009). This generates fear that ACTs will

become inefficient (Pradines et al., 2010).

Artemisinins are normally metabolized into DHA which is their active compound

(Wongsrichanalai et al., 1999; Mwai et al., 2009). It is through this compound that artemisinins

exert their effect to malarial parasites. Furthermore, in aqueous media artemisinins are broken

down to dihydroartemisinin. Thus artemisinins have similar IC50 values (Wongsrichanalai et al.,

1999).

DHA with a mean IC50 of 1.861 nM (95% CI 1.489-2.233 nM) (Table 3), was the most

active drug (Table 4 and Figure 7); all parasites being susceptible to the lowest IC50 value

among all the drugs that were tested (Table 5 and Figure 6) in line with Mwai et al., (2009). It

gave significant statistical difference in the in vitro activities with all the other drugs except PQ

and CQ (Table 6).

Artemisinins have a short half-life and target all the stages of P. falciparum having much

more effect on the ring stages as compared to other drugs (Sompob et al., 2011). They are able to

reduce the parasite number up to as much as 10000 in one cycle (Woodrow et al., 2005). This

makes them the most active (Schlitzer 2008) accounting for the rapid activity of DHA compared

with all other antimalarial drugs.

The ability of P. falciparum to develop resistance to antimalarials is normally due to gene

polymorphism in the target or transport molecules (White, 1999; Tärning 2007). But the

mechanism of resistance to artemisinins is uncertain and polymorphisms in several candidate

genes have been proposed (Eastman and Fidock, 2009; Gama et al. 2010), which include:

38

(i.) Single nucleotide polymorphism (SNPs) in pfatpase6, a gene which codes for a

SERCA-type calcium-translocating ATPase (Jambou et al., 2005; Eastman and Fidock,

2009; Gama et al., 2010).

(ii.) SNPs or amplification in codons 86, 184, 1034, 1042 and 1246 of pfmdr1, which

codes for P. falciparum multidrug resistance (PfMDR1) (Cecilia et al., 2004; Price et al.,

2004; Sidhu et al., 2006; Eastman and Fidock, 2009).

(iii.) SNPs in codons 74, 75, 76, 220, 271, 326 and 371 of pfcrt which codes for P.

falciparum chloroquine resistance transporter (PfCRT) (Cecilia et al., 2004)

(iv.) Polymorphisms in the gene coding for P. falciparum multidrug resistance –

associated protein (PfMRP) (Raj et al., 2009).

PfMDR1 and PfCRT are transport proteins located on the membrane of the DV of the

parasite, and since this is one of the proposed active site of artemisinins, it thus controls drug

accretion (Eastman and Fidock, 2009) as any mutation in these genes results in a modified

protein which impairs drug accumulation and consequently their potencies (Valderramos and

Fidock, 2006).

Kilifi isolate have been characterised to have one mutation and copy number of pfmdr1

(Mwai et al., 2009) further explaining the high activity of DHA. Pfmdr1 amplification is not

commonly found in Africa (Holmgren et al., 2006; Ursing, et al., 2006; Sisowath et al., 2007;

Mwai et al., 2009) due to low usage of MFQ (Mwai et al., 2009).

Additionally, as drug pressure contributes greatly in promoting the development and

spread of drug resistance due to their short elimination life (Schlitzer 2008), artemisinins exert

little drug pressure (Wongsrichanalai et al., 1999) consequently reducing the chances of

resistance, hence the observed high activity towards all the isolates.

Despite the lack of demonstrate able artemisinins resistance there are reports of delayed

clearance times (Noedl et al., 2008; Dondorp et al., 2009; Witkowski et al., 2010) whereby

parasites survive in presence of artemisinins longer times as would be expected but later get

killed. As such, as opposed to the classical resistant phenotype this points out a different

resistance mechanism (Witkowski et al., 2010). Thus reduced artemisinins response in the ring-

stage could provide an answer to the reports of resistance in Cambodia (Benoit-Vical et al.,

2007; Witkowski et al., 2010) whereby a proportion of rings remain dormant in presence of the

drug and resume growth after withdrawal of the drug (Sompob et al. 2011). In other words, P.

39

falciparum uses a quiescence mechanism that allows it to survive ART treatment (Witkowski et

al., 2010). Indeed, even though artemisinins are still effective resistance will eventually emerge

(Yavo et al., 2010).

When the IC50s of two antimalarials are positively correlated, this indicates cross

resistance (Yavo et al., 2010). However, the positive correlation in the in vitro activities of DHA

and other antimalarials, except for MFQ, was insignificant reassuring the use of ACTs as first

line antimalarial drugs. The significant correlation with MFQ can be attributed to technical

failures that resulted in reduced MFQ response. Despite this observation, polymorphisms and

amplification of pfmdr1 indeed does reduce DHA and MFQ sensitivity (Pickard et al., 2003;

Reed et al., 2004; Sidhu et al., 2005; Gama et al., 2010). As the causation of this polymorphism

is the usage of MFQ, which is rare in Africa and Kenya indeed, in addition to the observed low

pfmdr1 copy number and polymorphism in Kilifi isolates (Mwai et al., 2009), this correlation is

highly unlikely. Based on this observation of unlikely MFQ reduced response, it is also unlikely

for the observed correlation between MFQ and PYR. Indeed, they have different modes of

action, PYR, an antifolate, inhibits dihydrofolate reductase in the folate biosynthetic pathway

while MFQ, an arylaminoalcohol, inhibits heme digestion (Schlitzer et al., 2008).

Unexpectedly CQ demonstrated a higher IC50 value second to DHA (Table 4 and

figure 13). With a mean IC50 of 24.29 nM (95% CI 13.2-35.46 nM) all isolates were sensitive to

CQ (Table 3, 5 and figure 12) in line with Mwai et al., (2009). But based on the lower IC50

cutoff value, one isolate was resistant to CQ. There was no significant statistical difference

between its in vitro activities as compared to the other antimalarials except PYR (Table 6). This

demonstrates that CQ is regaining its activities in this part of Kenya after its withdrawal.

Just as in the case of artemisinins, PfMDR1 and PfCRT control accumulation of CQ and

consequently the pH in the DV. Hence they are involved in chloroquine resistance too, whereby

polymorphisms in pfmdr1 and pfcrt have been found in CQ resistant parasites (Cecilia et al.,

2004; Gama et al., 2010). Infact, the response to CQ is much reduced when polymorphisms in

both genes are present (Cecilia et al., 2004). In a study on Kilifi isolates only one Pfmdr1 and the

pfcrt mutation in codon 76 were present in Kilifi isolates (Mwai et al., 2009). This explains the

low levels of CQ resistance observed in the isolates in addition to the reduced CQ drug pressure

since its ban more than a decade ago.

40

PQ was also very active; with a mean IC50 value of 30.34 nM (95% CI 26.96-33.72 nM)

all isolates were sensitive to it in line with another study of isolates from Kilifi (Mwai et al.,

2009). Similar results were obtained for isolates from other malaria endemic areas (Deloron et

al., 1985; Barends et al., 2007; Basco and Ringwald 2003). Like CQ there was no significant

statistical difference between its in vitro activities as compared to the other antimalarials except

PYR.

PQ is a CQ based antimalarial consisting of two CQ molecules and is used in

combination with DHA in the ACT, Artekin, which is a possible substitute of Coartem

(Karunajeewa et al., 2008; Yeka et al., 2008; Thanh et al., 2009, Mwai et al., 2009).

Polymorphisms in pfcrt- 76 and pfmdr1- 86 don’t contribute to piperaquine resistance despite the

contribution of pfcrt to reduced response to piperaquine (Muangnoicharoen et al., 2009; Mwai et

al., 2009).

As piperaquine is effective against CQ resistant parasites (Basco and Ringwald 2003;

Mwai et al 2009) and with the current observed regaining of CQ sensitivity, this predicts

development of resistance to it (Karunajeewa et al., 2008; Muangnoicharoen et al., 2009; Mwai

et al., 2009). As a result of this, the effectiveness of PQ in areas of high CQ resistance e.g. the

South east of Asia will be short lived as compared to Africa (Karunajeewa et al., 2008; Yeka et

al., 2008; Thanh et al., 2009; Mwai et al., 2009).

Amodiaquine is only suggested for treatment of falciparum malaria (WHO 2000; Sasi et

al., 2009) due to its toxicity (Hatton et al., 1986; Neftel et al., 1986; Sasi et al., 2009). As it

demonstrated effectiveness against CQ resistant parasites, which was prevalent, amodiaquine use

was more around the 1990s (Fadat et al., 1991; Sasi et al., 2009). And due to this, it has been

adopted for CQ replacement in most African countries or better still as second line.

Consequently, it is a commonly used drug in Africa (WHO 2007; Sasi et al., 2009). On the same

note, it has been used in Kenya as a second line drug between 1998 and 2008 (Shretta et al.,

2000; Sasi et al., 2009) and as a front line between 2004 and 2006 (Zurovac 2008; Sasi et al.,

2009).

The use of ACTs which combines amodiaquine results in resistance to

monodesethylamodiaquine which predicts resistance to amodiaquine (Nawaz et al., 2009).

41

Further emphasizing the need for a new and different antimalarial (Bruno et al., 2010 a).

However, despite some of the isolates from Kilifi being resistant to DE-A and MFQ, 12.5% and

62.5% respectively they were still more active than LMF and Q (These had 100% sensitivity).

The mean IC50 of DE-A was 37.8 nM (19.46-56.14 nM), more active following PQ. Like CQ

and PQ there was no significant statistical difference between its in vitro activities as compared

to the other antimalarials except PYR.

DE-A is the active compound of AQ and having a similar structure as CQ, it also

inhibits haem detoxification (Legrand et al., 2008; Eastman and Fidock 2009) and it has efficacy

towards CQ-resistant parasites (Olliaro and Mussano 2003; Eastman and Fidock, 2009).

Polymorphisms in pfcrt and pfmdr1 are associated with its resistance (Bray1996; Ochong 2003;

Fitch 2004; Dokomajilar et al., 2006; Mwai et al., 2009)

Furthermore, AQ has been the second line of treatment in Kenya since SP was introduced in

1999 and has remained so until now. This drug has partially been used in several sites in Kenya

(Sidhu et al., 2002; Mwai et al 2009), including Kilifi, even before the withdrawal of CQ (Sidhu

et al., 2005; Mwai et al 2009).

The observed high DE-A sensitivity results from high CQ sensitivity (Kokwaro et al.,

2007; Kobbe et al., 2008) and the similar chemical structure between amodiaquine and

chloroquine. Furthermore, there has been less amodiaquine drug pressure in Kenya as it has been

always used as a backup drug (Sidhu et al., 2002; Sidhu et al., 2005; Mwai et al 2009).

MFQ had a mean IC50 of 41.7 nM (95% CI 23.64-59.75 nM). Except DHA it had no

significant statistical difference between its in vitro activities as compared to the other

antimalarials.

Polymorphisms in pfcrt-76 and pfnhe (a sodium hydrogen exchanger gene) are not

associated with MFQ resistance (Price et al., 2004; Okombo et al., 2010). While polymorphisms

and amplification of pfmdr1 reduce MFQ sensitivity (Pickard et al., 2003; Reed et al., 2004;

Sidhu et al., 2005; Gama et al., 2010). Indeed pfmdr1 amplification mainly determines MFQ

susceptibility (Pukrittayakamee et al., 2000; Price et al., 2004; Okombo et al., 2010).

Despite the uncommon use of MFQ compared to other antimalarial drugs in Africa

(Okombo et al., 2010) and Kilifi County for that matter, 62.5% MFQ-resistant isolates were

42

observed in this study. This observation is in contrast to another study on isolates from Kilifi

County whereby the isolates were found to be sensitive to MFQ (Okombo et al., 2010).

Furthermore, the isolates were found to have 1 copy of the pfmdr1 gene (Mwai et al., 2009),

explaining the susceptibility of parasites to MFQ.

A more plausible explanation for this difference can be attributed to a greater disparity

in the threshold IC50 for MFQ resistance. Some literatures cites > 30 nM (Ndong et al., 2003;

Bruno et al., 2010), others > 40 nM (Hatabu et al., 2010) and > 50 nM (Randrianarivelojosia et

al., 2004). But still based on the previous two IC50 cutoff values, a high MFQ resistance was

observed, with 62.5% (5 out of 8 isolates) and 50% (4 out of 8 isolates) resistant, respectively.

Thus the observation of a high failure of MFQ sensitivity to isolates from Kilifi is

highly unlikely and may be attributed to technical failures. Or perhaps, the presence of isolates

that are inherently resistant to MFQ could help to explain this observation (Gari-Toussaint et al.,

2002; Henry et al, 2006; Yavo et al., 2010). However, there is a possibility of MFQ resistance as

it can be in rodent malaria (Sidhu et al., 2005, 2006). And if this were to happen in P.

falciparum, it would complicate the fight against malaria (Gama et al. 2010).

In Kenya, currently Coartem is the front line drug (Kokwaro et al., 2007; Mwai et al

2009). But LMF usage results in resistance to Coartem (Dokomajilar et al., 2006; Humphreys et

al., 2007; Happi et al., 2009; Mwai et al 2009). And bearing in mind the reported artemisinins

resistance in South East Asia, it’s predictable that this drug will lose its efficacy (Dondorp et al .,

2009; Mwai et al., 2009).

LMF was more active than Q and PYR following MFQ. With a mean IC50 of 46.5

nM (95% CI 31.40-61.60 nM), all (100%) isolates were sensitive to it in line with another study

on Kilifi field isolates (Mwai et al., 2009) and studies in other endemic areas (Anderson et al.,

2005; Basco and Ringwald 2007; Kaddouri et al., 2008; Mayxay et al., 2007, Parola et al., 2007;

Pradines et al., 2006; Mwai et al., 2009). Like MFQ it had no significant statistical difference

between its in vitro activities as compared to the other antimalarials except DHA. It had positive

correlation with all the drugs except CQ and Q. This inverse relationship between the activities

of LM and CQ is in line with Mwai et al. studies suggesting that selection of LM resistance

would be associated with an increase in CQ activity (2009).

43

Parasites with wild type pfmdr1 and pfcrt are less sensitive to LM and on the contrary,

mutants of these genes are resistant to CQ (Dokomajilar et al., 2006; Humphreys et al., 2007;

Happi et al., 2009; Sisowath et al., 2005, 2007, 2009; Mwai et al., 2009). This helps to explain

the inverse relationship between CQ and LM activity (Pradines et al., 1999, Price et al., 2006;

Mwai et al., 2009). However, at the moment Coartem is still effective (Falade et al., 2008 a,b;

Kobbe et al., 2008; Yeka, et al., 2008; Mwai et al., 2009).

For the treatment of complicated malaria and CQ resistance, Q is the recommended drug

(Ndong et al., 2003).

When compared to the other antimalarial drugs Q was among the least active drugs

despite 100% sensitivity among the isolates. But it was more active than PYR. This total

sensitivity is in line with other studies (Je ˆ rome et al., 2003; Djaman et al., 2004; Toure et al.,

2008; Yavo et al., 2010). Other studies carried out in the coastal region of Kenya including Kilifi

showed a similar Q activity (Watkins et al., 1987; Pasvol et al., 1991: Haruki et al., 1998;

Okombo et al., 2010). And so as from other parts of Kenya and other African countries (Ndong

et al., 2003; Odhiambo and Odulaja 2005; Agnamey et al., 2006; Pradines et al., 2006; Tinto et

al., 2006; Quashie et al., 2007; Okombo et al., 2010). With a mean IC50 value of 46.7 nM (95%

CI 36.78-56.62 nM) and like MFQ and LMF it had no significant statistical difference between

its in vitro activities as compared to the other antimalarials except DHA.

Polymorphisms in pfmdr1 and pfcrt as well as pfmdr1 amplification are associated with

Q resistance (Pickard et al., 2003; Reed et al., 2003; Lakshmanan et al., 2005; Sidhu et al., 2005

Nsobya et al., 2010; Chaijaroenkul et al., 2010).

Polymorphism in pfnhe modify the in vitro response to Q whereby an increase in the

number of DNNND repeats (1–5) in ms4760 microsatellite is associated with less sensitivity to

Q (Ferdig et al., 2004; Henry et al., 2009; Okombo et al., 2010) and discrepancy in its copy

number occur (Vinayak et al., 2007; Okombo et al., 2010).

In a study of 29 isolates from Kilifi characterised them to have 1- 3 DNNND (Aspartic

acid-asparagine- asparagine-asparagine-aspartic acid) repeats. 17 and 6 isolates had 2 and 1

repeats, respectively, and the remaining 6 isolates had 3 repeats. The increase from 1 to 2 repeats

was associated with decreasing susceptibility to Q, and the change from 2 to 3 repeats rendered

44

parasites more susceptible (Okombo et al., 2010). However, another study shows no decrease in

Q activity in parasites with 3 DNNND repeats (Henry et al., 2009; Okombo et al., 2010).

Pfmdr1 is an important contributing factor in Q resistance and the isolates have been

characterised to have pfmdr1-86 mutation and 1 pfmdr1 copy number (Mwai et al., 2009;

Okombo et al., 2010). The potency of Q can be altered by the degree of accumulation inside the

DV, which is the site of haem detoxification (Valderramos and Fidock, 2006). The low number

of DNNND repeats and the low rate of Pfmdr1 polymorphism could explain the high activity of

QN, in addition to less drug pressure due to correct use (Bustos et al., 1994; Okombo et al.,

2010).

There is lack of acquiescence in the correct use of Q (Fungladda et al., 1998; Okombo

et al., 2010) resulting in less drug pressure and hence resistance not developing towards it, and

further explaining the observed high Q activity. Thus QN is still effective (Okombo et al., 2010).

Unfortunately there are accounts of Q resistance (Brasseur et al., 1988; Kilimali et al.,

1990; Mutanda 1999; Pettinelli et al., 2004; Okombo et al., 2010) and reduced response

(Pukrittayakamee et al., 1994, 2000; Parola et al., 2001; Roche, et al., 2003; Molinier et al.,

2004; Adam et al., 2004; Achan et al., 2009; (Okombo et al., 2010). Which even if are however,

based on effectiveness studies and that it is possible it could be out of the lack of acquiescence in

correct Q use (Okombo et al., 2010), but still it would be better to undertake resistance

surveillance of the same (Tinto et al., 2001; Okombo et al., 2010). To emphasize on this, drug

pressure resulting from the usage of Q selects resistant parasites (Randrianarivelojosia et al.,

2004; Henry et al., 2006; Pradines et al., 2006; Tinto et al., 2006; Parola et al., 2007; Yavo et al.,

2010).

PYR was the least active drug, with a mean IC50 value of 18074 nM (95% CI 9483-

26665 nM) all isolates were resistant to it. Surprisingly, except PQ, DHA, CQ and DE-A it had

no significant statistical difference between its in vitro activities as compared to the other

antimalarials.

PYR resistance is associated with mutations in the dihydrofolate reductase (DHFR) gene

dhfr, whereby the mutations at codon 108 (Serine-108-Asparagine) leads to PYR resistance, with

45

level increasing when additional mutations, Asparagine-51-Isoleucine and Cystine-59-Arginine,

occurs. This becomes even higher with the Isoleucine-164-Leucine mutation (Kiara et al., 2009).

Infact, mutations in dhfr are the major determinant of resistance towards Pyrimethamine-

Sulfadoxine (PYR-SD), which occurs as a triple mutant involving the mutations; (Serine-108-

Asparagine, Asparagine-51-Isoleucine, and Cystine-59-Arginine) (Nzila 2006; Kiara et al.,

2009). And the addition of the mutation Isoleucine-164-Leucine (quadruple mutant parasites)

leads to higher resistance of the same (Wilairatana et al., 1997; Watkins et al., 1997; Kiara et al.,

2009). This mutation is indeed found in Africa (Hame et al., 2008; Kiara et al., 2009), no wonder

the high PYR-SD resistance.

Polymorphism in Guanine triphosphates (GTP)-cyclohydrolase also leads to PYR-SD

resistance, which is also an enzyme in the folate metabolism pathway (Nair et al., 2008; Kiara et

al., 2009).

The above dhfr gene mutations have been documented in isolates from Kilifi (Kiara et

al., 2009) explaining the resistance towards PYR.

Besides, there was resistance towards PYR-SD in Kilifi County as early as the mid-1990s

(Shretta et al., 2000; Kiara et al., 2009) which proved futile in early 2000s and its use was

eventually banned in 2004 (Amin et al., 2007; Kiara et al., 209).

Despite its ban, there is still usage of PYR-SD (Clarke et al., 2008; Temperley et al.,

2008; Kiara et al., 2009) and based on its affordability its abused leading to drug pressure

(Wichmann et al., 2003; Ochong et al., 2003 b; Parola et al., 2007; Kiara et al., 2009) and

consequently the observed high PYR resistance (Kiara et al., 2009). Therefore, the use of PYR-

SD should be highly regulated (Spalding et al., 2010).

46

CHAPTER FIVE

CONCLUSIONS

Artemisinins are still active in Kilifi County. They are more active than other standard

antimalarials. No cross-resistance was observed with other antimalarials. Isolates from initial and

recurrent malaria have similar in vitro response to artemisinins and other antimalarials.

RECOMMENDATIONS

One; continued public awareness should be done to encourage vector and parasite control

to help fight malaria. Two; to avoid drug pressure, effective and correct use of antimalarials

should be reinforced to avoid resistance emergence. Three; in addition to the need of more

studies on polymorphisms of the antimalarials candidate genes in isolates from Kilifi County

more studies should be carried out in other parts of Kenya and other endemic countries to

provide unremitting surveillance of response to artemisinins. This will be useful in providing

information that will advice the making of treatment policy. Four; understanding how

artemisinins act and the resistance mechanisms as well as development of new and different

antimalarials to avoid relying only on artemisinins will help protect artemisinins against

resistance. Five; hopes in the fight against malaria rely on an effective and acceptable vaccine.

Thus the search towards such development should be accelerated.

47

REFFERENCES

Achan J., Tibenderana J., K., Kyabayinze D., Wabwire Mangen F., Kamya M. R., Dorsey G.,

D’Alessandro U., Rosenthal P. J., and A. O. Talisuna. (2009). Effectiveness of quinine

versus artemether-lumefantrine for treating uncomplicated falciparum malaria in

Ugandan children: randomized trial. British Medical Journal 339: 2763.

Adam I., Ali D. M., Noureldien W., and Elbashir M. I. (2005). Quinine for the treatment of

chloroquine-resistant Plasmodium falciparum malaria in pregnant and non-pregnant

Sudanese women. Annals of Tropical Medicine and Parasitology 99: 427–429.

Agnamey P., Brasseur P., de Pecoulas P. E., Vaillant M., and Olliaro P. (2006). Plasmodium

falciparum in vitro susceptibility to antimalarial drugs in Casamance (southwestern

Senegal) during the first 5 years of routine use of artesunate-amodiaquine. Antimicrobial

Agents and Chemotherapy 50: 1531–1534.

Aide P., Bassat Q. and Alonso P.L. (2007). Towards an effective malaria vaccine. Archives of

Disease in Childhood 92: 476-479.

Amin A. A., Zurovac D., Kangwana B. B., Greenfield J., Otieno D. N., Akhwale W. S., and

Snow R. W. (2007). The challenges of changing national malaria drug policy to a

rtemisinin-based combinations in Kenya. Malaria Journal 6: 72.

Anderson T.J., Nair S., Qin H., Singlam S., Brockman A., Paiphun L., Nosten F. (2005). Are

transporter genes other than the chloroquine resistance locus (pfcrt) and multidrug

resistance gene (pfmdr) associated with antimalarial drug resistance? Antimicrobial

Agents and Chemotherapy 49: 2180-2188.

Asawamahasakda W., Ittarat I., Pu Y.M., Ziffer H., Meshnick S.R. (1994). Reaction of

antimalarialendoperoxides with specific parasite proteins. Antimicrobial Agents and

Chemotherapy 38: 1854–1858.

Auerbach T. (2002). Antibiotics targeting ribosomes: crystallographic studies. Current Drug

Targets. Infectious Disorders 2: 169–186.

48

Bacon D.J, Latour C., Lucas C., Colina O., Ringwald P. and Picot S. (2007). Comparison of a

SYBR Green I-Based Assay with a Histidine-Rich Protein II Enzyme-Linked

Immunosorbent Assay for In VitroAntimalarial Drug Efficacy Testing and Application to

Clinical Isolates. Antimicrobial Agents and Chemotherapy 51: 1172–1178.

Barends M., Jaidee A., Khaohirun N., Singhasivanon P. and Nosten F. (2007). In vitro activity of

ferroquine (SSR 97193) against Plasmodium falciparum isolates from the Thai-Burmese

border. Malaria Journal 6: 81.

Basco L.K. and Ringwald P. (2003). In vitro activities of piperaquine andother 4-

aminoquinolines against clinical isolates of Plasmodium falciparum in Cameroon.

Antimicrobial Agents and Chemotherapy 47: 1391–1394.

Basco L.K. and Ringwald P. (2007). Molecular epidemiology of malaria in Cameroon. XXIV.

Trends of in vitro antimalarial drug responses in Yaounde, Cameroon. The American

Journal of Tropical Medicine and Hygiene 76: 20–26.

Bennett T. N., Paguio M., Gligorijevic B., Seudieu C., Kosar A. D., Davidson E. and Roepe P.

D. (2004). Novel, rapid, and inexpensive cell-based quantification of antimalarial drug

efficacy. Antimicrobial Agents and Chemotherapy 48: 1807–1810.

Benoit-Vical F., Lelievre J., Berry A., Deymier C., Dechy-Cabaret O., Cazelles J., Loup C.,

Robert A., Magnaval J. and Meunier B. (2007). Trioxaquines:new antimalarial agents

active on all erythrocytic forms including gametocytes. Antimicrobial Agents and

Chemotherapy 51: 1463–1472.

Brasseur P., Kouamouo J., Brandicourt O., Moyou-Somo R. and Druilhe P. (1988). Patterns of in

vitro resistance to chloroquine, quinine, and mefloquine of Plasmodium falciparum in

Cameroon, 1985–1986. The American Journal of Tropical Medicine and Hygiene 39:

166–172.

Bray P.G., Hawley S.R., Mungthin M., Ward S.A. (1996). Physicochemical properties correlated

with drug resistance and the reversal of drug resistance in Plasmodium falciparum.

Molecular Pharmacology 50: 1559–1566.

49

Breman J.G., Alilio M.S. and Mills A. (2004). Conquering the intolerable burden of malaria:

what’s new, what’s needed: a summary. The American Journal of Tropical Medicine and

Hygiene 71: 1–15.

Bruno P., Sébastien B., Maud H., Claude O., Eric B., Rémy A.,Eric D. and Christophe R. (2010).

Absence of association between pyronaridine in vitro responses and polymorphisms in

genes involved in quinoline resistance in Plasmodium falciparum. Malaria Journal 9: 1-

7. (a)

Bruno P., Thierry P., Khaled E., Sébastien B., Lionel B., Marie-Catherine R., Daniel P., Pascal

M., Christophe R.,and Denis M. (2010). Quinine-Resistant Malaria in Traveler Returning

from Senegal, 2007. Emerging Infectious Disease 16: 546-548. (b)

Bustos M.D.G., Gay P.F. and B. Diquet. (1994). In-vitro tests on Philippine isolates of

Plasmodium falciparum against four standard antimalarials and four qinghaosu

derivatives. Bulletin of the World Health Organization 72: 729-735

Chaijaroenkul W., Wisedpanichkij R. and Na-Bangchang K. (2010). Monitoring of in vitro

susceptibilities and molecular markers of resistance of Plasmodium falciparum isolates

from Thai-Myanmar border to chloroquine, quinine, mefloquine and artesunate. Acta

Tropica 113: 190–194.

Clarke S.E., Jukes M.C., Njagi J. K., Khasakhala L., Cundill B., Otido J., Crudder C., Estambale

B. B., and Brooker S. (2008). Effect of intermittent preventive treatment of malaria on

health and education in schoolchildren: a cluster-randomised, double-blind, placebo-

controlled trial. Lancet 372: 127–138.

Craig A. and Scherf A. (2001). Molecules on the surface of the Plasmodium falciparum infected

erythrocyte and their role in malaria pathogenesis and immune evasion. Molecular &

Biochemical Parasitology 115: 129–143.

Cui L and Su X. (2009). Discovery, mechanisms of action and combination therapy of

artemisinin. Expert Review of Anti-Infective Therapy 7: 999–1013.

50

Day K.P. and Marsh K. (1991). Naturally acquired immunity to Plasmodium falciparum.

Immunology Today 12: 68-71.

Deloron P., Le Bras J., Ramanamirija J. A. and Coulanges P. (1985). Plasmodium falciparum in

Madagascar: in vivo and in vitro sensitivity to seven drugs. Annals of Tropical Medicine

and Parasitology 79, 357–365.

Denis M.B., Tsuyuoka R., Lim P., Lindegardh N., Yi P., Top S.N., Socheat D., Fandeur T.,

Annerberg A., Christophel E.M. and Ringwald P. (2006). Efficacy of artemether-

lumefantrine for the treatment of uncomplicated falciparum malaria in northwest

Cambodia. Tropical Medicine and International Health 11: 1800-1807.

Djaman J., Abouanou S., Basco L. and Kone M. (2004). Limits of the efficacy of chloroquine

and sulfadoxine-pyrimethamine in Northern Abidjan (Cote d'Ivoire): Combined in vivo

and in vitro studies. Sante 14: 205–209.

Dokomajilar C., Nsobya S.L., Greenhouse B., Rosenthal P.J. and Dorsey G. (2006). Selection of

Plasmodium falciparum pfmdr1 alleles following therapy with artemether-lumefantrine in

an area of Uganda where malaria is highly endemic. Antimicrobial Agents and

Chemotherapy 50: 1893–1895.

Dondorp A.M., Nosten F., Yi P., Das D., Phyo A.P., Tarning J., Lwin K.M., Ariey F.,

Hanpithakpong W., Lee S.J., Ringwald P., Silamut K., Imwong M., Chotivanich K., Lim

P., Herdman T., An S.S., Yeung S., Singhasivanon P., Day N.P., Lindegardh N., Socheat

D., White N.J. (2009). Artemisinin resistance in Plasmodium falciparum malaria. The

New England Journal of Medicine 361: 455-467.

Druile P., Moreno A., Blanc C., Brasseur P.H. and Jacquier P. (2001). A colometric in vitro drug

sensitivity assay for Plasmodium based on a highly sensitive double site lactate

dehydrogenase antigen-capture Elisa. The American Journal of Tropical Medicine and

Hygiene 64: 233–241.

Eastman R.T. and Fidock D.A. (2009). Artemisinin-based combination therapies: a vital tool in

efforts to eliminate malaria. Nature Reviews. Microbiology 7: 864–874.

51

Fadat G., Le Bras J., Hengy C., Louis J.P., Gimou M.M. and Verdier F. (1991). Efficacy of

amodiaquine against chloroquine-resistant malaria in Cameroon. Lancet 338: 1092.

Falade C.O., Ogundele A.O., Yusuf B.O., Ademowo O.G. and Ladipo S.M. (2008). High

efficacy of two artemisinin-based combinations (artemether- lumefantrine and artesunate

plus amodiaquine) for acute uncomplicated malaria in Ibadan, Nigeria. Tropical Medicine

and International Health 13: 635–643. (a)

Falade C.O., Ogunkunle O.O., Dada-Adegbola H.O., Falade A.G., Ibarra de Palacios P., Hunt P.,

Virtanen M., Oduola A. M. and Salako L. A. (2008). Evaluation of the efficacy and

safety of artemether-lumefantrine in the treatment of acute uncomplicated Plasmodium

falciparum malaria in Nigerian infants and children. Malaria Journal 7: 246. (b)

Feachem R. and Sabot O. (2008). A new global malaria eradication strategy. Lancet 371: 1633–

1635.

Ferdig M.T., Cooper R.A., Mu J., Deng B., Joy D.A., Su X.Z. and Wellems T.E. (2004).

Dissecting the loci of low-level quinine resistance in malaria parasites. Molecular

Microbiology 52: 985–997.

Fitch C.D. (2004). Ferriprotoporphyrin IX, phospholipids and the antimalarial actions of

quinoline drugs. Life Sciences 74: 1957–1972.

Fungladda, W., Honrado E. R., Thimasarn K., Kitayaporn D., Karbwang J., Kamolratanakul P.

and Masngammueng R. (1998). Compliance with artesunate and quinine + tetracycline

treatment of uncomplicated falciparum malaria in Thailand. Bulleting of the World

Health Organisation 76: Supplementary 1:59-66.

Gama B.E., Almeida de Oliveira N.K., de Souza J.M, Santos F., de Carvalho L.M.J., Melo

Y.F.C., Rosenthal F.P., Daniel-Ribeiro C.T. and Ferreira-da-Cruz M.F. (2010). Brazilian

Plasmodium falciparum isolates:investigation of candidate polymorphisms for

artemisinin resistance before introduction ofartemisinin-based combination therapy.

Malaria Journal 9: 1-5.

52

Gardner M.J., Shallom S.J., Carlton J.M., Salzberg S.L., Nene V., Shoaibi A., Ciecko A., Lynn

J., Rizzo M., Weaver B., Jarrahi B., Brenner M., Parvizi B., Tallon L., Moazzez A.,

Granger D., Fujii C.,Hansen C., Pederson J., Feldblyum T., Peterson J., Suh B. and

Angiuoli S. (2002). Sequence of Plasmodium falciparum chromosomes 2, 10, 11 and 14.

Nature 419: 531-534.

Gari-Toussaint M., Pradines B., Mondain V., Keundjian A., Dellamonica P., Le Fichoux Y.

(2002). Senegal and malaria. True prophylactic failure of mefloquine. Presse Medicale

31: 1136

Gogtay N.J., Kadam V.S, Karnad D.R., Kanbur A., Kamtekar K.D. and Kshirsagar N.A. (2000).

Probable resistance to parenteral artemether in Plasmodium falciparum: case reports from

Mumbai (Bombay), India. Annals of Tropical Medicine and Parasitoogy 94: 519–520.

Greenwood B.M., Bojang K., Whitty C.J. and Targett G.A. (2005). Malaria. Lancet 365: 487–

498.

Gregson A. and Plowe C.V. (2005). Mechanisms of resistance of malaria parasites to antifolates.

Pharmacological Reviews 57: 117–145.

Griffith K.S., Lewis L.S., Mali S. and Parise M.E. (2007). Treatment of malaria in the United

States: a systematic review. JAMA: the journal of american medical association 297:

2264-2277.

Happi C.T., Gbotosho G.O., Folarin O.A., Sowunmi A., Hudson T., O’Neil M., Milhous W.,

Wirth D.F. and Oduola A.M.. (2009). Selection of Plasmodium falciparum multidrug

resistance gene 1 alleles in asexual stages and gametocytes by artemether-lumefantrine in

Nigerian children with uncomplicated falciparum malaria. Antimicrobial Agents and

Chemotherapy 53: 888– 895.

Hatton C.S., Peto T.E. and Bunch C. (1986). Frequency of severe neutropenia associated with

amodiaquine prophylaxis against malaria. Lancet 1: 411– 414.

Hartwig C.L., Rosenthal A.S., D'Angelo J., Griffin C.E., Posner G.H., Cooper R.A. (2009).

Accumulation of artemisinin trioxane derivatives within neutral lipids of Plasmodium

53

falciparum malaria parasites is endoperoxide-dependent. Biochemical Pharmacology 77:

322-336.

Haruki K., Winstanley P. A., Watkins W. M. and Marsh K. (1998). Quinine sensitivity of

isolates of Plasmodium falciparum from the coast of Kenya. Transactions of the Royal

Society of Tropical Medicine and Hygiene 92:195–196.

Hastings I. M., Paget-McNicol S. and Saul A. (2004). Can mutation and selection explain

virulence in human P.falciparum infections? Malaria Journal 3: 2.

Hastings I.M. and Watkins W.M. (2005). Intensity of malaria transmission and the evolution of

drug resistance. Acta Tropica 94: 218–229.

Hay S.I., Rogers D.J., Toomer J.F. and Snow R.W. (2000). Annual Plasmodium falciparum

entomological inoculation rates (EIR) across Africa: literature survey, internet access and

review. Transactions of the Royal Society of Tropical Medicine and Hygiene 94: 113–

127.

Hatabu T., Kawazu S., Kojima S., Singhasivanon P., Krudsood S., Looareesuwan S. and Kano S. (2004). A pilot field trialof an in vitro drug suscpetibility test using the anaeropack malaria culture system on the Thai-Myanmar border. Tropical medicine and Hygiene 32: 335-337.

Hay S.I., Guerra C.A., Gething P.W., Patil A.P., Tatem A.J., Noor A.M., Kabaria C.W., Manh

B.H., Elyazar I.R., Brooker S., Smith D.L., Moyeed R.A. and Snow R.W. (2009). A

world malaria map: Plasmodium falciparum endemicity in 2007. PLoS Medicine 6:

e1000048.

Henry M., Diallo I. and Bordes J., Ka S., Pradines B., Diatta B., M'Baye P.S., Sane M., Thiam

M., Gueye P.M., Wade B., Touze J.E., Debonne J.M., Rogier C. and Fusai T. (2006).

Urban malaria in Dakar, Senegal: chemosusceptibility and genetic diversity of

Plasmodium falciparum isolates. The American Journal of Tropical Medicine and

Hygiene 75: 146–151.

Henry M., Briolant S., Zettor A., Pelleau S., Baragatti M., Baret E., Mosnier J., Amalvict R.,

Fusai T., Rogier C. and Pradines B. (2009). Plasmodium falciparum Na/Hexchanger 1

54

transporter is involved in reduced susceptibility to quinine. Antimicrobial Agents and

Chemotherapy 53: 1926– 1930.

Holmgren G., Bjorkman A., and Gil J. P. (2006). Amodiaquine resistance is not related to rare

findings of pfmdr1 gene amplifications in Kenya. Tropical Medicine and Inernational

Health 11: 1808–1812.

Humphreys G. S., Merinopoulos I., Ahmed J., Whitty C. J., Mutabingwa T. K., Sutherland C. J.

and Hallett R. L. (2007). Amodiaquine and artemether- lumefantrine select distinct alleles

of the Plasmodium falciparum mdr1 gene in Tanzanian children treated for

uncomplicated malaria. Antimicrobial Agents and Chemotherapy 51: 991–997.

Jambou R., Legrand E., Niang M., Khim N., Lim P., Volney B., Ekala M.T., Bouchier C.,

Esterre P., Fandeur T. and Mercereau-Puijalon O. (2005). Resistance of Plasmodium

falciparum field isolates to in-vitro artemether and point mutations of the SERCA-type

PfATPase6. Lancet 366: 1960-1963.

Janeway C., Travers P., Walport M. and Schlomnik M. J. (2005). Immuno Biology: The Immune

System in Health and Disease. 6th Ed. New York: Garland Science Publishing.

Jensen J. B. (2002). In Vitro Culture of Plasmodium Parasites. In D. L. Doolan, Malaria Methods

and Protocols (pp. 477-488). Totowa, NJ: Humana Press, Inc.,.

Johnson J.D., Dennull R.A., Gerena L., Lopez-Sanchez M., Roncal N. E. and Waters N.C.

(2007). Assessment and Continued Validation of the Malaria SYBR Green I-Based

Fluorescence Assay for Use in Malaria Drug Screening. Antimicrobial Agents and

Chemotherapy 51: 1926–1933.

Kaddouri H., Nakache S., Houzé S., Mentré F. and Le Bras J. (2006). Assessment of the Drug

Susceptibility of Plasmodium falciparum Clinical Isolates from Africa by Using a

Plasmodium Lactate DehydrogenaseImmunodetection Assay and an Inhibitory Maximum

Effect Model for Precise Measurement of the 50-Percent Inhibitory Conc. Antimicrobial

Agents and Chemotherapy 33: 3343–3349.

55

Kaddouri H., Djimde A., Dama S., Kodio A., Tekete M., Hubert V., Kone A., Maiga H., Yattara

O., Fofana B., Sidibe B., Sangare C.P., Doumbo O. and Le Bras J. (2008). Baseline in

vitro efficacy of ACT component drugs on Plasmodium falciparum clinical isolates from

Mali. International Journal of Parasitology 38: 791–798.

Kannan R., Kumar K., Sahal D., Kukreti S. and Chauhan V.S. (2005). Reaction of artemisinin

with haemoglobin:implications for antimalarial activity. The Biochemical Journal 385:

409–418.

Karunajeewa H.A., Mueller I., Senn M., Lin E., Law I., Gomorrai P.S., Oa O., Griffin S., Kotab

K., Suano P., Tarongka N., Ura A., Lautu D., Page-Sharp M., Wong R., Salman S., Siba

P., Ilett K. F. and Davis T.M. (2008). A trial of combination antimalarial therapies in

children from Papua New Guinea. The New England Journal of Medicine 359: 2545–

2557.

Kiara S.M., Okombo J., Masseno V., Mwai L., Ochola I., Borrmann S. and Nzila A. (2009). In

Vitro Activity of Antifolate and Polymorphism in Dihydrofolate Reductase of

Plasmodium falciparum Isolates from the Kenyan Coast: Emergence of Parasites with

Ile-164-Leu Mutation. Antimicrobial Agents and Chemotherapy 53: 3793–3798.

Kilimali V. A. (1990). The in vitro response of Plasmodium falciparum to amodiaquine, quinine

and quinidine in Tanga region, Tanzania. East Africa Medical Journal 67: 336–340.

Klayman D.L. (1985). Qinghaosu (artemisinin): an antimalarial drug from China. Science 228:

1049–1055.

Kobbe R., Klein P., Adjei S., Amemasor S., Thompson W. N., Heidemann H., Nielsen M.V.,

Vohwinkel J., Hogan B., Kreuels B., Buhrlen M., Loag W., Ansong D. and May J.

(2008). A randomized trial on effectiveness of artemether-lumefantrine versus artesunate

plus amodiaquine for unsupervised treatment of uncomplicated Plasmodium falciparum

malaria in Ghanaian children. Malaria Journal 7: 261.

Kokwaro G., Mwai L. and Nzila A. (2007). Artemether/lumefantrine in the treatment of

uncomplicated falciparum malaria. Expert Opinion on Pharmacotherapy 8: 75–94.

56

Krishna S., Bustamante L., Haynes R.K. and Staines H. M. (2008). Artemisinins: their growing

importance in medicine. Trends in Pharmacological Sciences 29: 520–527.

Krungkrai S.R. and Yuthavong Y. (1987). The antimalarial action on Plasmodium falciparum of

qinghaosu and artesunate in combination with agents which modulate oxidant stress.

Transactions of the Royal Society of Tropical Medicine and Hygiene 81: 710–714.

Lakshmanan V., Bray P.G., Verdier-Pinard D., Johnson D.J., Horrocks P., Muhle R.A., Alakpa

G.E., Hughes R.H., Ward S.A., Krogstad D.J., Sidhu A.B. and Fidock D.A. (2005). A

critical role for PfCRT K76T in Plasmodium falciparum verapamil-reversible

chloroquine resistance. The EMBO Journal 24: 2294–2305.

Le Bras J., Musset L. and Clain J. (2006). Antimalarial drug resistance. Medecine Maladies

Infectieuses 36: 401-405.

Legrand E., Volney B., Meynard J.B., Mercereau-Puijalon O., Esterre P. (2008). In vitro

monitoring of Plasmodium falciparum drug resistance in French Guiana: a synopsis of

continuous assessment from 1994 to 2005. Antimicrobial Agents and Chemotherapy 52:

288–298.

Leonardo K.B. and Pascal R. (2003). In Vitro Activities of Piperaquine and Other 4-

Aminoquinolines against Clinical Isolates of Plasmodium falciparum in Cameroon.

Antimicrobial Agents and Chemotherapy 47: 1391–1394.

Lim I. and Macfadden G.I. (2010). The evolution, metabolism and functions of the apicoplast.

Philosophical Transactions of the Royal Society of london 365: 749-763.

Luxemburger C., Brockman A., Silamut K., Nosten F., van Vugt M., Gimenez F.,

Chongsuphajaisiddhi T. and White N.J. (1998). Two patients with falciparum malaria and

poor in vivo responses to artesunate. Transactions of the Royal Society Tropical Medicine

and Hygiene 92: 668-669.

Mayxay M., Barends M., Brockman A., Jaidee A., Nair S., Sudimack D., Pongvongsa T.,

Phompida S., Phetsouvanh R., Anderson T., White N. J. and Newton P.N. (2007). In vitro

antimalarial drug susceptibility and pfcrt mutation among fresh Plasmodium falciparum

57

isolates from the Lao PDR (Laos). The American Journal of Tropical Medicine and

Hygiene 76: 245–250.

Mbogo C.M., Mwangangi J.M., Nzovu J., Gu W., Yan G., Gunter J.T., Swalm C., Keating

J.,Regens J.L., Shililu J.I., Githure J.I. and Beier J.C. (2003). Spatial and temporal

heterogeneity of Anopheles mosquitoes and Plasmodium falciparum transmission along

the Kenyan coast. The American journal of tropical medicine and hygiene 68: 734-742.

Menard R. (2005). Medicine: knockout malaria vaccine? Nature 433: 113-114.

Miller J.M., Korenromp E.L., Nahlen B.L. and R W.S. (2007). Estimating the number of

insecticidetreatednets required by African households to reach continent-wide malaria

coverage targets. JAMA 297: 2241-2250.

Molinier S., Imbert P., Verrot D., Morillon M., Parzy D. and Touze J.E. (1994). Plasmodium

falciparum malaria: type R1 quinine resistance in East Africa. Presse Medicale 23: 1494.

Murray C. K. and Bennett J.W. (2009). Rapid Diagnosis of Malaria. Interdisciplinary

Perspectives on Infectious Disease 2009: 1- 7.

Muangnoicharoen S., Johnson D. J., Looareesuwan S., Krudsood S. and Ward S. A. (2009). Role

of known molecular markers of resistance in the antimalarial potency of piperaquine and

dihydroartemisinin in vitro. Antimicrobial Agents and Chemotherapy 53: 1362–1366.

Mutanda L. N. (1999). Assessment of drug resistance to the malaria parasite in residents of

Kampala, Uganda. East African Medicine Journal 76: 421–424.

Mwai L., Steven M. K., Abdi A., Lewa P., Anja R., Abdi D., Pete B., Kevin M., Steffen B. and

Alexis N. (2009). In Vitro Activities of Piperaquine, Lumefantrine, and

Dihydroartemisinin in Kenyan Plasmodium falciparum Isolates and Polymorphisms in

pfcrt and pfmdr1. Antimicrobial Agents and Chemotherapy 53: 5069–5073.

Nair, S., Miller B., Barends M., Jaidee A., Patel J., Mayxay M., Newton P., Nosten F., Ferdig M.

T. and Anderson T.J. (2008). Adaptive copy number evolution in malaria parasites. PLoS

Genetics 4:e1000243.

58

Nawaz F., Nsobya S.L., Kiggundu M., Joloba M., Rosenthal P.J. (2009). Selection of parasites

with diminished drug susceptibility by amodiaquine-containing antimalarial regimens in

Uganda. The Journal of Infectious Disease 200: 1650-1657.

Ndong J. M., Atteke C., Aubouy A., Bakary M., Lebibi J. and Deloron P. (2003). In vitro

activity of chloroquine, quinine, mefloquine and halofantrine against Gabonese isolates

of Plasmodium falciparum. Tropical Medicine and International Health 8: 25–29.

Neftel K.A., Woodtly W., Schmid M., Frick P.G. and Fehr J. (1986). Amodiaquine induced

agranulocytosis and liver damage. British Medical Journal 292: 721–723.

Noedl H., Attlmayr B., Wernsdorffer W. H, Kollaritsch H., and Miller S. (2004). A Histidine-

Rich Protein 2−Based Malaria Dug Sensitivity Assay For Field Use. The American

Journalof Tropical Medicine and Hygiene 71: 711–714.

Noedl H., Bronnert J., Yingyuen K., Attlmayr B., Kollaritsch H. and Fukuda M. (2005). Simple

Histidine-Rich Protein 2 Double-Site Sandwich Enzyme-Linked. Antimicrobial Agents

and Chemotherapy 35: 3575–3577.

Noedl H., Se Y., Schaecher K., Smith B.L., Socheat D. and Fukuda M.M. (2008). Evidence of

artemisinin-resistant malaria in western Cambodia. The New England Journal of

Medicine 359: 2619-2620.

Nosten F. and White N.J. (2007). Artemisinin-based combination treatment of falciparum

malaria. The American Journal of Tropical Medicine and Hygiene 77: 181–192.

Nsobya S.L., Kiggundu M., Nanyunja S., Joloba M., Greenhouse B. and Rosenthal P.J. (2010).

In vitro sensitivities of Plasmodium falciparum to different antimalarial drugs in Uganda.

Antimicrobial Agents and Chemotherapy 54: 1200–1206.

Nzila, A. (2006). The past, present and future of antifolates in the treatment of Plasmodium

falciparum infection. The Journal of Antimicrobial Chemotherapy 57: 1043–1054.

Ochong E.O., Broek v.d., Keus K. and Nzila A. (2003). Short report: association between

chloroquine and amodiaquine resistance and allelic variation in the Plasmodium

falciparum multiple drug resistance 1 gene and the chloroquine resistance transporter

59

gene in isolates from the upper Nile in southern Sudan. The American Journal of

Tropical Medicine and Hygiene 69: 184–187.

Ochong E., Nzila A., Kimani S., Kokwaro G., Mutabingwa T., Watkins W., and Marsh K.

(2003). Molecular monitoring of the Leu-164 mutation of dihydrofolate reductase in a

highly sulfadoxine/pyrimethamine-resistant area in Africa. Malaria Journal 2: 46.

Odhiambo R.A. and Odulaja A. (2005). Parasite lactate dehydrogenase assay for the

determination of antimalarial drug susceptibility of Kenyan field isolates. East African

Medical Journal 82: 118–122.

Oduola A.M., Sowunmi A., Milhous W.K., Kyle D.E., Martin R.K., Walker O. and Salako L.A.

(1992). Innate resistance to new antimalarial drugs in Plasmodium falciparum from

Nigeria. Transactions of the Royal Society and Tropical Medicine Hygiene 86: 123-126.

Okombo J., Kiara S. M., Rono J., Mwai L., Pole L., Ohuma E., Borrmann S., Ochola L.I. and

Nzila A. (2010). In Vitro Activities of Quinine and Other Antimalarials and pfnhe

Polymorphisms in Plasmodium Isolates from Kenya. Antimicrobial Agents and

Chemotherapy 54: 3302–3307

Olliaro P. and Mussano P. (2003). Amodiaquine for treating malaria. Cochrane Database System

Review CD000016.

O'Neill P.M., Barton V.E. and Ward S.A. (2010). The molecular mechanism of action of

artemisinin--the debate continues. Molecules 15: 1705-1721.

Parola P., Ranque S., Badiaga S., Niang M., Blin O., Charbit J. J., Delmont J. and Brouqui P.

(2001). Controlled trial of 3-day quinine-clindamycin treatment versus 7-day quinine

treatment for adult travelers with uncomplicated falciparum malaria imported from the

tropics. Antimicrobial Agents and Chemotherapy 45: 932–935.

Parola, P., Pradines B., Simon F., Carlotti M.P., Minodier P., Ranjeva M.P., Badiaga S., Bertaux

L., Delmont J., Morillon M., Silai R., Brouqui P. and Parzy D. (2007). Antimalarial drug

susceptibility and point mutations associated with drug resistance in 248 Plasmodium

falciparum isolates imported from Comoros to Marseille, France in 2004–2006. The

American Journal of Tropical Medicine and Hygiene 77: 431–437.

60

Pasvol G., Newton C. R.,Winstanley P. A., Watkins W. M., Peshu N. M., Were J. B., Marsh K.

and Warrell D. A. (1991). Quinine treatment of severe falciparum malaria in African

children: a randomized comparison of three regimens. The American Journal of Tropical

Medicine and Hygiene 45: 702–713.

Pettinelli F., M.E. Pettinelli M.E., Eldin de P.P., Millet J., Michel D., Brasseur P. and Druilhe P.

(2004). Short report: high prevalence of multidrug resistant Plasmodium falciparum

malaria in the French territory of Mayotte. The American Journal of Tropical Medicine

and Hygiene 70: 635–637.

Pickard A.L., Wongsrichanalai C., Purfield A., Kamwendo D., Emery K., Zalewski C.,

Kawamoto F., Miller R.S., Meshnick S.R. (2003). Resistance to antimalarials in

Southeast Asia and genetic polymorphisms in pfmdr1. Antimicrobial Agents and

Chemotherapy 47: 2418-2423.

Pradines B., Tall A., Fusai T., Spiegel A., Hienne R., Rogier C., Trape J. F., Le Bras J. and Parzy

D. (1999). In vitro activities of benflumetol against 158 Senegalese isolates of

Plasmodium falciparum in comparison with those of standard antimalarial drugs.

Antimicrobial Agents and Chemotherapy 43: 418–420.

Pradines B., Hovette P., Fusai T., Atanda H.L., Baret E., Cheval P., Mosnier J., Callec A., Cren

J., Amalvict R., Gardair J.P. and Rogier C. (2006). Prevalence of in vitro resistance to

eleven standard or new antimalarial drugs among Plasmodium falciparum isolates from

Pointe-Noire, Republic of the Congo. Journal of Clinical Microbiology 44: 2404–2408.

Pradines B., Briolant S., Henry M., Oeuvray C., Baret E., Amalvict R.,Didillon E., and Rogier C.

(2010). Absence of association between pyronaridine in vitro responses and

polymorphisms in genes involved in quinoline resistance in Plasmodium falciparum.

Malaria Journal 9: 339.

Price R.N., Uhlemann A.C., Brockman A., McGready R., Ashley E., Phaipun L., Patel R., Laing

K., Looareesuwan S., White N.J., Nosten F. and Krishna S. (2004). Mefloquine resistance

in Plasmodium falciparum and increased pfmdr1 gene copy number. Lancet 364: 438–

447.

61

Price, R.N., Uhlemann A.C., van Vugt M., Brockman A., Hutagalung R., Nair S., Nash D.,

Singhasivanon P., Anderson T.J., Krishna S., White N.J. and Nosten F. (2006). Molecular

and pharmacological determinants of the therapeutic response to artemether-lumefantrine

in multidrug-resistant Plasmodium falciparum malaria. Clinical Infectious Disease 42:

1570–1577.

Pukrittayakamee S., Supanaranond W., Looareesuwan S., Vanijanonta S. and White N.J. (1994).

Quinine in severe falciparum malaria: evidence of declining efficacy in Thailand.

Transactions of the Royal Society Tropical Medicine and Hygiene 88: 324–327.

Pukrittayakamee S., Viravan C., Charoenlarp P.,Yeamput C., Wilson R.J. and White N.J.

(1994). Antimalarial effects of rifampin in Plasmodium vivax malaria. Antimicrobial

Agentsand Chemotherapy 38: 511-514.

Pukrittayakamee S., Chantra A., Vanijanonta S., Clemens R., Looareesuwan S. and White N.J.

(2000). Therapeutic responses to quinine and clindamycin in multidrug-resistant

falciparum malaria. Antimicrobial Agents and Chemotherapy 44: 2395–2398.

Quashie N.B., Duah N.O., Abuaku B. and Koram K.A. (2007). The in-vitro susceptibilities of

Ghanaian Plasmodium falciparum to antimalarial drugs. Annals of Tropical Medicine and

Parasitology 101: 391–398.

Randrianarivelojosia M., Randrianasolo L., Randriamanantena A., Ratsimbasoa A., Rakotoson

J.D. (2004). Susceptibility of Plasmodium falciparum to the drugs used to treat severe

malaria (quinine) and to prevent malaria (mefloquine, cycloguanil) in Comoros Union

and Madagascar. South African Medical Journal 94: 47–51.

Raj D.K., Mu J., Jiang H., Kabat J., Singh S., Sullivan M., Fay M.P., McCutchan T.F. and Su

X.Z. (2009). Disruption of a Plasmodium falciparum multidrug resistance-associated

protein (PfMRP) alters its fitness and transport of antimalarial drugs and glutathione.

Journal of Biological Chemistry 284: 7687-7696.

Randrianarivelojosia M., Raharimalala L.A., Randrianasolo L., Ratsimbasoa A., Rason M.A.,

Ariey F., Jambou R. (2001). Madagascan isolates of Plasmodium falciparum showing

62

low sensitivity to artemether in vitro. Annals of Tropical Medicine and Parasitology 95:

237-43.

Randrianarivelojosia M., Randrianasolo L., Randremanana R. V., Randriamanantena A., Ratsimbasoa A., Rakotoson J.D. (2004). Susceptibility of Plasmodium falciparum to the drugs used to treat severe malaria (quinine) and to prevent malaria (mefloquine, cycloguanil) in Comoros Union and Madagascar. South Africa Medical Journal 94: 47-51.

Reed M.B., Saliba K.J, Caruana S.R., Kirk K., Cowman A.F. (2000). Pgh1 modulates sensitivity

and resistance to multiple antimalarials in Plasmodium falciparum. Nature 403: 906–909.

Roche J., Guerra-Neira A., Raso J. and Benito A. (2003). Surveillance of in vivo resistance of

Plasmodium falciparum to antimalarial drugs from 1992 to 1999 in Malabo (Equatorial

Guinea). The American Journal of Tropical Medicine and Hygiene 68: 598–601.

Ro D.K., Paradise E.M., Ouellet M., Fisher K.J., Newman K.L., Ndungu J.M., Ho K.A., Eachus

R.A., Ham T.S., Kirby J., Chang M.C., Withers S.T., Shiba Y., Sarpong R. and Keasling

J.D. (2006). Production of the antimalarial drug precursor artemisinic acid in engineered

yeast. Nature 440: 940-943.

Rogers W.O., Sem R., Tero T., Chim P., Lim P., Muth S., Socheat D., Ariey F. and

Wongsrichanalai C. (2009). Failure of artesunate-mefloquine combination therapy for

uncomplicated Plasmodium falciparum malaria in southern Cambodia. Malaria Journal

8: 10.

Roos D.S., Crawford M. J., Donald R. G. K, Fraunholz M., Harb O. S., He C. Y., Kissinger J. C.,

Shaw M. K. and Striepen B. (2002). Mining the Plasmodium genome database to define

organellar function: what does the apicoplast do? Phillosophical Transactions of the

Royal Society Biological Science 357: 35 - 46.

Roper C., Pearce R., Nair S., Sharp B., Nosten F. and Anderson T. (2004). Intercontinental

spread of pyrimethamine-resistant malaria. Science 305: 1124.

63

Sahr F., Willoughby V.R., Gbakima A.A. and Bockarie M.J. (2001). Apparent drug failure

following artesunate treatment of Plasmodium falciparum malaria in Freetown, Sierra

Leone: four case reports. Annals of Tropical Medicine and Parasitology 95: 445–449.

Sasi P., Abdulrahaman A, Mwai L., Muriithi S., Straimer J., Schieck E., Rippert A., Bashraheil

M., Salim A., Peshu J., Awuondo K., Lowe B., Pirmohamed M., Winstanley P., Ward S.,

Nzila A. and Borrmann S. (2009). In Vivo and In Vitro Efficacy of Amodiaquine against

Plasmodium falciparum in an Area of Continued Use of 4-Aminoquinolines in East

Africa. The Journal of Infectious Diease 199: 1575 – 1582.

Schlitzer M. (2008). Antimalarial Drugs – What is in Use and What is in the Pipeline. Archives

de pharmacy 341: 149 – 163.

Shretta R., Omumbo J., Rapuoda B. and Snow R.W. (2000). Using evidence to change

antimalarial drug policy in Kenya. Tropical Medicine and International Health 5: 755–

64.

Sidhu A.B., Verdier-Pinard D. and Fidock D.A. (2002). Chloroquine resistance in Plasmodium

falciparum malaria parasites conferred by pfcrt mutations. Science 298: 210–213.

Sidhu A.B., Valderramos S.G. and D.A. Fidock. (2005). pfmdr1 mutations contribute to quinine

resistance and enhance mefloquine and artemisinin sensitivity in Plasmodium falciparum.

Molecular Microbiology 57: 913–926.

Sidhu A.B., Uhlemann A.C., Valderramos S.G., Valderramos J.C., Krishna S. and Fidock D.A.

(2006). Decreasing pfmdr1 copy number in Plasmodium falciparum malaria heightens

susceptibility to mefloquine, lumefantrine, halofantrine, quinine, and artemisinin. Journal

of Infectious Disease 194: 528-535.

Sisowath C., Strömberg J., Mårtensson A., Msellem M., Obondo C., Björkman A., Gil J.P.

(2005). In vivo selection of Plasmodium falciparum pfmdr1 86N coding alleles by

artemether-lumefantrine (Coartem). Journal of Infectious Disease 191: 1014-1017.

Sisowath C., Ferreira P. E., Bustamante L. Y., Dahlstrom S., Martensson A., Bjorkman A.,

Krishna S., and Gil J. P. (2007). The role of pfmdr1 in Plasmodium falciparum tolerance

64

to artemether-lumefantrine in Africa. Tropical Medicine and International Health 12:

736–742.

Smilkstein M., Sriwilaijaroen N, Kelly J.X., Wilairat P. and Riscoe M. (2004). Simple and

Inexpensive Fluorescence-Based Technique for High-Throughput Antimalarial Drug

Screening. Antimicrobial Agents and Chemotherapy 48: 1803–1806.

Snounou G. and Renia L. (2007). The vaccine is dead--long live the vaccine. Trends in

Parasitology 23: 129-132.

Snow R.W, Guerra C.A, Noor A.M, Myint H.Y and Hay S.I. (2005). The global distribution of

clinical episodes of Plasmodium falciparum malaria. Nature 434: 214–217.

Sompob S., Wirichada P., Richard J. M, Sue J. L., Joel T., Niklas L., Kesinee C., François N.,

Nicholas P. J. D., Duong S., Nicholas J. W., Arjen M. D., and Lisa J. W. (2011). Intrahost

modeling of artemisinin resistance in Plasmodium falciparum. PNAS. 108: 397–402.

Spalding M.D., Eyase F.L., Akala H.M., Bedno S.A., Prigge S.T, Coldren R.L, Moss W.J. and

Waters N.C. (2010). Increased prevalence of the pfdhfr/phdhps quintuple mutant and

rapid emergence of pfdhps resistance mutations at codons 581 and 613 in Kisumu,

Kenya. Malaria Journal 9: 338

Straimer J. (2009). Studies on evolution of drug resistance in P. falciparum malaria in vivo and

in vitro. Doctor of natural sciences thesis, Heidelberg University, Germany 5-6.

Tärning J. (2007). Piperaquine; Bioanalysis, drug metabolism and pharmacokinetics. Göteborg,

Sweden: ISBN. 11-13

Taubes G. (2000). Searching for a parasite’s weak spot. Science 290: 434-437.

Thanh N.X., Trung T.N., Phong N.C., Thien N.X., Dai B., Shanks G.D., Chavchich M. and

Edstein M.D. (2009). Open label randomized comparison of dihydroartemisinin-

piperaquine and artesunate-amodiaquine for the treatment of uncomplicated Plasmodium

falciparum malaria in central Vietnam. Tropical Medicine and International Health 14:

504–511.

65

Temperley M., Mueller D.H., Njagi J.K., Akhwale W., Clarke S.E., Jukes M.C., Estambale B.B.,

and Brooker S. (2008). Costs and cost-effectiveness of delivering intermittent preventive

treatment through schools in western Kenya. Malaria Journal 7:196.

Tinto H., Ouedraogo J.B., Traore B., Coulibaly S.O., Guiguemde T.R. (2001). In vitro

susceptibility of 232 isolates of Plasmodium falciparum to antimalarials in Burkina Faso

(West Africa). Bulleting de la Societe de Pathologie Exotique. 94: 188–191

Tinto, H., C. Rwagacondo, C. Karema, D. Mupfasoni, W. Vandoren, E. Rusanganwa, A. Erhart,

C. Van Overmeir, E. Van Marck, and U. D’Alessandro. (2006). In-vitro susceptibility of

Plasmodium falciparum to monodesethylamodiaquine, dihydroartemisinin and quinine in

an area of high chloroquine resistance in Rwanda. Transactions of the Royal Society

Tropical Medicine and Hygiene 100: 509 –514.

Touré A.O., Koné L.P., Jambou R., Konan T.D., Demba S., Beugre G.E. and Koné M. (2008). In

vitro susceptibility of P. falciparum isolates from Abidjan (Côte d'Ivoire) to quinine,

artesunate and chloroquine. Sante. 18: 43-47.

Tripathi R.P., Mishra R.C., Dwivedi N., Tewari N. and Verma S.S. (2005). Current Status of

Malaria Control. Current Medicinal Chemistry 12: 2643-2659.

Tritten T., Matile T., Brun R. and Wittlin S. (2009). A new double-antibody sandwich ELISA

targeting Plasmodium. Malaria Journal 8: 226.

Ursing, J., Kofoed P. E., Rombo L. and Gil J. P. (2006). No pfmdr1 amplifications in samples

from Guinea-Bissau and Liberia collected between 1981 and 2004. Journal of Infectious

Disease 194: 716–718.

Valderramos S.G. and Fidock D. (2006). Transporters involved in resistance to antimalarial

drugs. Trends in Pharmacological Scence 27: 594–601.

Van Dooren G.G., Su V., D'Ombrain M.C., McFadden G.I. (2002). Processing of an Apicoplast

Leader Sequence in Plasmodium falciparum and the Identification of a Putative Leader

Cleavage Enzyme. Journal of Biological Chemistry 277: 23612-23619.

66

Vijaykadga S., Rojanawatsirivej C., Cholpol S., Phoungmanee D., Nakavej A. and

Wongsrichanalai C. (2006). In vivo sensitivity monitoring of mefloquine monotherapy

and artesunate-mefloquine combinations for the treatment of uncomplicated falciparum

malaria in Thailand in 2003. Tropical Medicine and International Health 11: 211-219.

Vinayak, S., M. T. Alam, M. Upadhyay, M. K. Das, V. Dev, N. Singh, A. P. Dash, and Y. D.

Sharma. (2007). Extensive genetic diversity in the Plasmodium falciparum Na/H

exchanger 1 transporter protein implicated in quinine resistance. Antimicrobial Agents

and Chemotherapy 51: 4508–4511.

Watkins W.M., Mberu E.K., Winstanley P.A. and Plowe C.V. (1997). The efficacy of antifolate

antimalarial combinations in Africa: a predictive model based on pharmacodynamic and

pharmacokinetic analyses. Parasitology Today 13: 459 – 464.

Wellems T.E. and Plowe C.V. (2001). Chloroquine-resistant malaria. Journal of Infectious

Disease 184: 770–776.

Wernsdorfer W.H. and Mc Gregor I. (1988). Malaria: Principles and Practice of Malariology .

London: Churchill Livingstone.

Watkins W. M., Howells R. E., Brandling-Bennett A. D., and Koech D. K. (1987). In vitro

susceptibility of Plasmodium falciparum isolates from Jilore, Kenya, to antimalarial

drugs. The American Journal of Tropical Medicine and Hygiene 37: 445–451.

White N. (1999). Antimalarial drug resistance and combination chemotherapy. Philosophical

Transactions of the Royal Societyof London 354: 739–749.

White N.J. (2001). Preventing antimalarial drug resistance through combinations. Drug

Resistance Updates 1: 3-9.

White N.J., Pongtavornpinyo W. (2003). The de novo selection of drug-resistant malaria

parasites. Proceedings of the Biological Sciences 270: 545–554.

White N.J. (2008). Qinghaosu (artemisinin): the price of success. Science 320: 330–334.

WHO. (2005). World malaria report 2005. Roll Back Malaria, World Health Organization.

67

Wichmann, O., T. Jelinek, G. Peyerl-Hoffmann, N. Muhlberger, M. P. Grobusch, J. Gascon, A.

Matteelli, C. Hatz, H. Laferl, M. Schulze, G. Burchard, S. da Cunha, J. Beran, P.

McWhinney, H. Kollaritsch, P. Kern, J. Cuadros, M. Alifrangis, and I. Gjorup. (2003).

Molecular surveillance of the antifolateresistant mutation I164L in imported African

isolates of Plasmodium falciparum in Europe: sentinel data from TropNetEurop. Malaria

Journal 2: 17.

Wilairatana, P., D. E. Kyle, S. Looareesuwan, K. Chinwongprom, S. Amradee, N. J. White, and

W. M. Watkins. (1997). Poor efficacy of antimalarial biguanide-dapsone combinations in

the treatment of acute, uncomplicated, falciparum malaria in Thailand. Annals of Tropical

Medicine and Parasitology 91: 125–132.

Witkowski B., Lelie`vre J., Barraga´n M.J.L., Laurent V., Su X., Berry A.and Benoit-Vical F.

(2010). Increased Tolerance to Artemisinin in Plasmodium falciparum Is Mediated by a

Quiescence Mechanism. Antimicrobial Agents and Chemotherapy 54: 1872–1877.

Wongsrichanalai C., Wimonwattrawatee T. , Sookto P., Laoboonchai A., Heppner D.G., Kyle

D.E. and Wernsdorfer W.H. (1999). In vitro sensitivity of Plasmodium falciparum to

artesunate in Thailand. Bulleting of the World Health Organisation 77: 392-398.

Wongsrichanalai C., Pickard A.L., Wernsdorfer W.H. and Meshnick S.R. (2002). Epidemiology

of drug-resistant malaria. Lancet 2: 209–218.

Woodrow C.J., Haynes R.K. and Krishna S. (2005). Artemisinins. Postgraduate Medical Journal

81: 71–78.

Wootton J.C., Feng X., Ferdig M.T., Cooper R.A., Mu J., Baruch D.I., Magill A.J. and Su X.Z.

(2002). Genetic diversity and chloroquine selective sweeps in Plasmodium falciparum.

Nature 418: 320-323.

World Health Organization (WHO). (2000). WHO Expert Committee on Malaria, twentieth

report. 30. Geneva: WHO.

World Health Organization (WHO). (2007). Antimalarial treatment policies. Vol 2008. Geneva:

WHO.

68

Wu Y. (2002). How might qinghaosu (artemisinin) and related compounds kill the

intraerythrocytic malaria parasite? A chemist’s view. Accounts of Chemical Research 35:

255–259.

Yang H., Liu D., Yang Y., Fan B., Yang P., Li X., Li C., Dong Y. and Yang C. (2003). Changes

in susceptibility of Plasmodium falciparum to artesunate in vitro in Yunnan Province,

China. Transactions of the Royal Society Tropical Medicine and Hygiene 97: 226-228.

Yavo W., K.B. Bla, A.J. Djaman, S.B. Assi, L.K. Basco, A. Mazabraud, and M. Koné. (2010). In

vitro susceptibility of Plasmodium falciparum to monodesethylamodiaquine, quinine,

mefloquine and halofantrine in Abidjan (Côte d'Ivoire). African Health Sciences 10: 111–

116.

Yeka A., Dorsey G., Kamya M.R., Talisuna A., Lugemwa M., Rwakimari J.B., Staedke S.G.,

Rosenthal P. J., Wabwire-Mangen F. and Bukirwa H. (2008). Artemether-lumefantrine

versus dihydroartemisinin-piperaquine for treating uncomplicated malaria: a randomized

trial to guide policy in Uganda. PLoS One 3: e2390.

Zurovac D., Njogu J., Akhwale W., Hamer D.H., Snow R.W. (2008). Translation of artemether-

lumefantrine treatment policy into paediatric clinical practice: an early experience from

Kenya. Tropical Medicine and International Health 13: 99–107.